EP3145540A2 - Chemically activated nanocapsid functionalized for cancer targeting - Google Patents

Chemically activated nanocapsid functionalized for cancer targeting

Info

Publication number
EP3145540A2
EP3145540A2 EP15796952.8A EP15796952A EP3145540A2 EP 3145540 A2 EP3145540 A2 EP 3145540A2 EP 15796952 A EP15796952 A EP 15796952A EP 3145540 A2 EP3145540 A2 EP 3145540A2
Authority
EP
European Patent Office
Prior art keywords
hev
capsid protein
vlp
modified capsid
cysteine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP15796952.8A
Other languages
German (de)
French (fr)
Other versions
EP3145540A4 (en
EP3145540B1 (en
Inventor
R. Holland Cheng
Li Xing
Chun Chieh Chen
Marie Christine STARK
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
University of California
Original Assignee
University of California
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by University of California filed Critical University of California
Publication of EP3145540A2 publication Critical patent/EP3145540A2/en
Publication of EP3145540A4 publication Critical patent/EP3145540A4/en
Application granted granted Critical
Publication of EP3145540B1 publication Critical patent/EP3145540B1/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5169Proteins, e.g. albumin, gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/525Virus
    • A61K2039/5258Virus-like particles
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/28011Hepeviridae
    • C12N2770/28111Hepevirus, e.g. hepatitis E virus
    • C12N2770/28122New viral proteins or individual genes, new structural or functional aspects of known viral proteins or genes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/28011Hepeviridae
    • C12N2770/28111Hepevirus, e.g. hepatitis E virus
    • C12N2770/28123Virus like particles [VLP]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2770/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses positive-sense
    • C12N2770/00011Details
    • C12N2770/28011Hepeviridae
    • C12N2770/28111Hepevirus, e.g. hepatitis E virus
    • C12N2770/28141Use of virus, viral particle or viral elements as a vector
    • C12N2770/28142Use of virus, viral particle or viral elements as a vector virus or viral particle as vehicle, e.g. encapsulating small organic molecule
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2810/00Vectors comprising a targeting moiety
    • C12N2810/40Vectors comprising a peptide as targeting moiety, e.g. a synthetic peptide, from undefined source
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N2333/00Assays involving biological materials from specific organisms or of a specific nature
    • G01N2333/005Assays involving biological materials from specific organisms or of a specific nature from viruses
    • G01N2333/08RNA viruses

Definitions

  • Vims-like particles can serve as nanocarriers for targeted delivery of diagnostics and therapeutics regimes, such as DNA/RNA and a variety of chemotherapeu ics.
  • Hepatitis E virus HEV
  • HEV vims-like particles are capsid protein ieosahedral cages that can be produced by expression of the major capsid protein HEV Open Reading Frame 2 (ORF2) in a eukaryotic expression system.
  • HEV VLPs are stable in acid and proteolytic
  • HEV VLPs represent a promising nano-carrier that can be exploited, e.g., for
  • the present invention provides a cysteine modified HEV V LP scaffold for altering HEV VLP surface functionality.
  • the present invention provides a modified capsid protein comprising a portion of hepatitis E virus (HEV) open Reading Frame 2 (ORF2) protein that is able to form an acid and proteolyticaliy stable HEV virus like particle (VLP), wherein: the portion of HEV ORF2 comprises a P-domain of the HEV ORF2 protein; the P-domain comprises at least one surface variable loop and a C -terminus; the P-domain comprises a cysteine in the at least one surface variable loop or at the C-termmus; and the HEV ORF2 portion retains its ability to form an acid and proteolyticaliy stable HEV VLP when the surface variable loop or C-terminal cysteine is chemically derivatized.
  • HEV hepatitis E virus
  • ORF2 open Reading Frame 2
  • VLP acid and proteolyticaliy stable HEV virus like particle
  • the HEV ORF2 portion retains its ability to form an acid and proteolyticaliy stable HEV VLP when the surface variable loop or C-terminal cysteine is alkylated, acylated, arylated, succinylated, oxidized, or conjugated to a detectable label or bioactive agent.
  • the modified capsid protein comprises an amino acid sequence at least 90%, 95%, or 99% identical, or identical, to residues 112-608 of the HEV ORE 2 protein of SEQ ID NO: 1, 2, 3, 4, 5, or 6.
  • the at least one P- domain surface variable loop or C-terminal cysteine is conjugated to a detectable label.
  • the detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
  • the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanocluster.
  • the at least one P-domain surface variable loop or C-terminai cysteine is conjugated to a bioactive agent.
  • the bioactive agent is a heterologous peptide.
  • the heterologous peptide is a ceil targeting ligand.
  • the cell targeting ligand is a cancer cell targeting liganc!. In some embodiments, the cancer ceil targeting ligand is LXY30. In some embodiments, the cancer cell targeting ligand is an antibody that binds an antigen expressed on the surface of a cancer cell. In some embodiments, the modified capsid protein further comprises a second cysteine in a P-domain surface variable loop or at the C-termimis of the P-domam. In some embodiments, the second cysteine is conjugated to a chemotherapeutic.
  • the second cysteine is conjugated to a detectable label.
  • the detectable label conjugated to the second cysteine comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomi c number greater than 20.
  • the detectable label conjugated to the second cysteine comprising the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanoc luster.
  • the at least one P-domain surface variable loop cysteine is alkylated, acylated, arylated, succinylated, or oxidized.
  • the at least one P-domain surface variable loop cysteine of HEV ORF2 replaces Y 85, T489, S533, N573, or T586 of HEV ORF2 or the C-terminal cysteine replaces residue 608 of HEV O F2.
  • the modified capsid protein is a component of an acid and
  • the acid and proteolytically stable HEV V LP encapsulates a bioactive agent.
  • the encapsulated bioactive agent is a heterologous nucleic acid, a heterologous peptide, a detectable label, a non-protemogenic amino acid, an oligosaccharide, a synthetic macromolecule, or a chemotherapeutic.
  • the encapsulated detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
  • the present invention provides a composition comprising the modified capsid protein of any one of the foregoing aspects or embodiments and a pharmaceutically acceptable excipient.
  • the composition comprises an HEV VLP having at least one cysteine within an HEV ORF2 P-domain surface variable loop or C-terminus that is chemically conjugated to a cell targeting ligand, a bioactive agent, or a detectable label. The chemical conjugation can result in a heterologous arrangement between the cysteine and the cell targeting ligand, bioactive agent, or detectable label.
  • the present invention provides a nucleic acid (e.g., isolated nucleic acid) comprising a polynucleotide sequence encoding any one of the foregoing modified capsid proteins.
  • the present invention provides an expression cassette comprising a promoter (e.g., heterologous promoter) operably linked to a polynucleotide sequence encoding an one of the foregoing modi fied capsid proteins.
  • the present invention provides a cell (e.g. , isolated cell or host cell) comprising the foregoing nucleic acid or the foregoing expression cassette.
  • the present invention provides a cell (e.g., isolated cell or host cell) comprising any one of the foregoing modified capsid proteins.
  • a cell e.g., isolated cell or host cell
  • the present invention provides an organism comprising any one of the foregoing modified capsid proteins.
  • the present invention provides a method of producing a modified capsid protein comprising cultivating any one of the foregoing cells, isolated cells, or host cells under conditions suitable to permit expression of the modified capsid protein.
  • the method further comprises purifying the capsid protein.
  • the method further comprises derivatizing the at least one P-domain surface variable loop or C-terminal cysteine.
  • the derivatizing comprises acylating, alkylating, arylating, succinylating, or oxidizing the P-domain surface variable loop or C-terminal cysteine.
  • the derivatizing comprises conjugating a bioactive agent to the at least one P-domain surface variable loop or C-terminal cysteine.
  • the present invention provides a method of directing an HE V VLP to a target cell comprising contacting a cell with the HEV-VLP, wherein the HEV VLP comprises any one of the foregoing modified capsid proteins, wherein the HEV VLP further comprises a cell targeting moiety having affinity for the target cell conjugated to the at least one P-domain surface variable loop or C-terminal cysteine, in some embodiments, the HE V VLP further comprises a detectable label, and the method further comprises detecting the detectable label.
  • the detecting the detectable label comprises detecting a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
  • the detecting the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises detecting a gold nanocluster.
  • the cell targeting moiety having affinity for the target cell conjugated to the at least one P-domain surface variable loop or C-terminal cysteine is a heterologous peptide comprising a cancer cell targeting ligarsd, and the target cell is a cancer cell.
  • the HEV VLP or a component thereof enters the cancer cell.
  • the HEV V I J' encapsulates a hioactive agent, and the method comprises delivering the bioactive agent to the intracellular region of the cell.
  • the encapsulated bioactive agent is a heterologous nucleic acid, a heterologous peptide, a detectable label, a non-proteinogenic amino acid, an oligosaccharide, a synthetic
  • the encapsulated detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
  • the encapsulated cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises an encapsulated gold nanocluster.
  • Figure 1 (A) Depicts the location of surface accessible flexible coil regions (P- domain surface variable loops) of HEV ORF 2, and residues Y485, T489, S533, N573, and T586, which lie within such flexible coil regions and were selected for cysteine mutation. (B) All the selected residues for cysteine mutation were located at the surface of the P- domain, with residue Y485, T489, T586 on the outmost surface, and residues S533 and N573 at side facing the VLP 5-fold axis.
  • FIG. 2 Characterization of chimeric HEV VLP carrying a cysteine replacement mutation.
  • A The cysteine mutation at 573 appeared to allow efficient biotinylation of the chimeric VLP as compared to the other mutation sites, i.e., Y485, T489, S533, and T586, although those mutations exhibited similar biotinylation after VLP disassembly
  • B Both the wild type VLP and the chimeric VLP had similar binding affinity to monoclonal antibody HEP40-4B, while chimeric VLP exhibited decreased reactivity to monoclonal antibody HEP230.
  • Figure 3 (A) The three-dimensional density map of HEV wt-VLP in complex with Fab fragments of HEP230 is displayed as isosurface along a 5-fold axis, showing extra density (arrow) in the gap of two 5-fold related protruding dimers. (B) The density agreed with the footprin t of the Fab molecule. (C) Residue N573 (sphere mode) is located next to the binding site of HEP230 that (D) allows only one Fab molecule binding per 5-fold region.
  • FIG. 4 The schematic of the two-steps chemical conjugation process including a thiol selective reaction and a Copper Catalyzed Azide-Alkyne Cycloaddition (CuAAC) or "click chemistry" reaction to form LXY30-VLPs.
  • CuAAC Copper Catalyzed Azide-Alkyne Cycloaddition
  • Method 1 (A) The Copper Catalyzed Azide-Alkyne Cycloaddition (CuAAC) reaction between Mal-Azide and Alkyne-LXY3() to form maleimide linked LXY30 (Mat- LXY30) was done first.
  • Method 2 (A) The conjugation process starting by labeling maleimide linked azide (Mal-Azide) at Cys sites of N573C-VLPs through Thiol selective-reaction to build azide linked VLPs (Azide-VLPs), followed by adding LXY30- Alkyne, ascorbic acid and CuS04 to form LXY30 linked VLPs (LXY30-VLPs).
  • FIG. 1 Damaged or dis-assembled LXY30-VLPs resulting from the conjugation process, due to the presence of reactive agents including azide, ascorbic acid and CuS04, can be separated from intact VLPs by, e.g., centrifugation, size exclusion chromatography, and the like.
  • Figure 5 In vitro fluorescence data of VLP-Cy5.5 and LXY30-VLP-Cy5.5 binding to MDA-MB-231 breast cancer ceils.
  • FIG. 6 The LXY30 chemical structure and its binding to U-87MG cells. Both scrambled LXY30 and LXY30 were directly conjugated to FITC, incubated with U-87MG cells, and detected by flow cytometry. Compared with the negative control, LXY30 showed obvious binding to U-87MG cells, which highly express alpha3-betal-integrin. The scrambled LXY30 showed minimum uptake due to non-specific staining from the fluorophore FiTC.
  • Figure 7 In vivo NIR fluorescence images of real-time tumor targeting
  • Forward 5 '-3' and Reverse 3 '-5 ' primers were designed with a cysteine coding sequence to the replace parental amino acid, a-e represent Y485C, T489C, S533C, N573C, and T586C respectively.
  • J Figure 9 Depicts the location of the surface variable loops and C -terminal residues of the P-domain of the HEV ORF2 capsid protein.
  • A Depicts a schematic diagram of P- domain surface variable loops of HEV ORF 2 capsid protein.
  • B Depicts the location of the P-domain surface variable loop residues and C-terminal residue on an HEV VLP.
  • One or more cysteines can be positioned within the indicated loop or C-terminal residues for conjugation to a molecular payload.
  • FIG. 10 Schematic depicting the application of chimeric HEV ORF2 VLPs as nanocarriers for drug delivery.
  • the periodicity of the capsid, the presence of surface- accessible amino acids with reactive moieties, and the tolerance of a single chain version of the capsid protein dimer to diverse peptide insertions enable dense, repetitive display of targeting iigands either by chemical conjugation or genetic insertion, and display of aptamers, glycoproteins, etc., by chemical conjugation.
  • HEV ORF2 VLPs also possess relatively large interior volumes that can be loaded with a variety of materials, including DNA plasmids. In comparison to other virus-like particles, HEV ORF2 VLPs possess strong structural plasticity, with a stability switch sensitive to calcium concentration.
  • FIG. 11 Cys-VLP conjugation to sulfur protected Auio 2 .
  • A Depicts two methods for covalent gold conjugation to Cys-VLP.
  • B Cys-VLP direct ligand exchange with Auio 2 (pMBA) 4 4.
  • C Cys-VLP maieimide conjugation with Au ⁇ -Ce-Maleimide.
  • D Cryo-EM Cys-VLP with A io2(pMBA) 4 a d the processed 3-D reconstruction,
  • Figure ⁇ 2 Schematic depicting the process used to encapsulate quantum dots in an HEV ORF2 VLP (top). Transmission electron microscope image of ferrite packaged VLPs showing darker intensity compared to package-free VLPs. DETAILED DESCRIPTION OF THE INVENTION I. Definitions
  • Hepatitis E virus refers to a vims, virus type, or virus class, which i) causes water-borne, infectious hepatitis; it) is distinguished from hepatitis A virus (HAV), hepatitis B virus (HBV), hepatitis C vims (HCV), or hepatitis D vims (HDV) in terms of serological characteristics: and iii) contains a genomic region that is homologous to a 1.33 kb cD ' NA inserted in pTZ Fl (ET1.1), a plasmid embodied in a E. coli strain deposited in American Type Culture Collection (ATCC) with accession number 67717.
  • HAV hepatitis A virus
  • HBV hepatitis B virus
  • HCV hepatitis C vims
  • HDV hepatitis D vims
  • capsid protein and “modified capsid protein,” with reference to HEV, refer to a mature or modified (e.g. , truncated, recombinantly mutated, or chemically derivatized) HEV open reading from 2 (ORF2) polypeptide.
  • ORF2 polypeptides or proteins refer to include the full-length polypeptide, and fragments thereof, and also include any substitutions, deletions, or insertions or other modifications made to the ORF2 proteins.
  • the capsid proteins must he capable of forming a virus like particle (VLP).
  • VLP virus like particle
  • the capsid protein contains at least residues 112-608 of HEV ORF2, although the capsid protein can tolerate various additional substitutions, deletions, or insertions so long as they are tolerated without abrogating VLP formation.
  • the term "modified capsid protein” refers to a capsid protein, or portion thereof, in which a surface variable loop of the P-domain of HEV ORF2 is modified to incorporate one or more cysteines that are not otherwise present in the wild-type capsid protein sequence.
  • modified capsid protein refers to a capsid protein, or portion thereof, in which the C-terminus (e.g., position 608) of HEV ORF2 is modified to incorporate one or more cysteines that are not otherwise present in the wild-type capsid protein sequence.
  • modified capsid protein refers to a capsid protein, or portion thereof, in which a cysteine (e.g. , a cysteine of a surface variable loop of the P-domain of H EV ORE 2 or a cysteine recombinantly introduced at position 608) is chemically derivatized to covalentlv conjugate to the protein at least one heterologous atom or molecule.
  • the cysteine can be inserted such that the HEV ORF2 protein length is increased, or the cysteine can replace one or more residues of a P-domain surface variable loop and/or C-terminus.
  • modified capsid proteins retain the ability to form HEV VLPs.
  • the one or more cysteines are conjugated to a bioactive agent (e.g., a cell-targeting ligand such as the peptide LXY30).
  • P-domain surface variable loops include one or more of , e.g., residues 475-493; residues 502-535; residues 539-569; residues 572-579; and residues 581-595 of HEV ORF 2 (SEQ ID NO: l, 2, 3, 4, 5, or 6).
  • P-domain surface variable loops further include the residues of polypeptides comprising an amino acid sequence that is at least about 80%, 85%, 90%, 95%, 99%, or more identical to one or more of SEQ ID NOS:l, 2, 3, 4, 5, or 6 and that correspond to one or more of residues 475-493; residues 502-535; residues 539-569; residues 572-579; and residues 581 -595 of SEQ ID NOS: l, 2, 3, 4, 5, or 6.
  • VLP virus-l ike particle
  • VLP refers to an icosahedrai shell (e.g., Tl or T 3) formed by a capsid protein.
  • VLPs are not infectious due to the lack of a viral genome.
  • VLP refers to a nonreplicating icosahedrai viral shell, derived from hepatitis E vims capsid protein HE V ORF2, a portion thereof. VLPs can form spontaneously upon recombinant expression of the protein in an appropriate expression system.
  • the VLP is formed from a modified capsid protein, e.g., a capsid protein containing one or more cysteine residues in a surface variable loop of HEV ORF2, or a portion thereof.
  • An HEV VLP can contain a mixture of modified and/or unmodified HEV ORF2 proteins.
  • an acid e.g., pH of, or of about, 6, 5.5, 5, 4.5, 4,
  • a population of HEV VLPs (e.g., modified or unmodified) can be incubated under acid and/or proteolytic conditions for a suitable period of time (e.g., for at least, or for at least about, 1 , 2, 3, 4, 5, 10, 15, 20, 30, 45, or 60 minutes) and then tested to determine the extent of quaternary structure retention.
  • a suitable period of time e.g., for at least, or for at least about, 1 , 2, 3, 4, 5, 10, 15, 20, 30, 45, or 60 minutes
  • HEV VLP refers to a modified HEV VLP that when incubated as a population of VLPs under acid and/or proteolytic conditions and assayed by electron microscopy, at least 10%, 25%, 50%, 75%, 90%, 95%, 99%, or 100% of the VLPs of the population retain their quaternary structure.
  • the HEV VLP can be delivered to a subject via an oral route and the efficiency of delivery assessed by detecting and/or quantifying: (i) an immune response to an antigen within the HEV VLP; (ii) a detectable label conjugated to, recombinantly introduced into, or encapsulated by the HEV V LP; or (iii) a biological response due to delivery to a cell of a bioactive agent associated with (e.g., recombinantly introduced into, conjugated to, or encapsulated by) the HEV VLP.
  • a bioactive agent associated with e.g., recombinantly introduced into, conjugated to, or encapsulated by
  • an acid and proteolytical!y stable modified HEV VLP refers to a modified HEV VLP that retains at least 10%, 25%, 50%, 75%, 90%, 95%, 99%, or 100%) of the oral delivery efficacy and/or cell entry activity of an unmodified HEV V LP.
  • heterologous refers to the two elements such as nucleic acids (e.g. , promoter or protein encoding sequence) or proteins (e.g. , an HEV ORF2 protein, or portion thereof, or modified capsid protein and another protein) that are not naturally found in the same relative positions.
  • a heterologous promoter of a gene refers to a promoter that is not naturally operably linked to that gene.
  • Hepatitis E virus is known to cause severe acute liver failure.
  • HEV belongs to the genus Hepevirus in the family Hepeviridae.
  • HEV contains a single-stranded positive- sense R.NA molecule of approximately 7.2-kb.
  • the RNA is 3' polyadenylated and includes three open reading frames (ORE).
  • ORF1 encodes viral nonstructural proteins, located in the 5' half of the genome.
  • ORF2 encodes a protein-forming viral capsid, located at the 3' terminus of the genome.
  • ORF3 encodes a 13.5-kDa protem, overlapped with C-terminus of ORF1 and N-terminus of ORF2.
  • ORF3 is associated with the membrane as well as with the
  • encapsulation refers to the envelopment of a heterologous substance, such as a heterologous nucleic acid or protein, a chemotherapeutic, an imaging agent, a ferrite nanoparticle etc., within the VLPs defined herein.
  • bioactive agent refers to any agent, drug, compound, or mixture thereof that targets a specific biological location (targeting agent) and/or provides some local or systemic physiological or pharmacologic effect that can be demonstrated in vivo or in vitro.
  • Non-limiting examples include drugs, hormones, vaccines, antibodies, antibody fragments, vitamins and co factors, polysaccharides, carbohydrates, steroids, lipids, fats, proteins, peptides, polypeptides, nucleotides, oligonucleotides, polynucleotides, and nucleic acids (e.g., mRNA, tRNA, snR A, R Ai, DNA, cDNA, antisense constructs, ribozymes, etc.).
  • a "pharmaceutically acceptable” or “pharmacologically acceptable” material is one that is not biologicall harmful or otherwise undesirable, i.e., the material may be
  • excipient refers to any essentially accessory substance that may be present in the finished dosage form of the composition of this invention.
  • excipient includes vehicles, binders, disintegrates, fillers (diluents), lubricants, glidants (flow enhancers), compression aids, colors, sweeteners, preservatives,
  • suspending/dispersing agents film formers/coatings, flavors and printing inks.
  • adjuvant refers to a compound that, when administered in conjunction with an antigen, augments the immune response to the antigen, but does not generate an immune response to the antigen when administered alone.
  • adjuvants can augment an immune response by several mechanism including lymphocyte recruitment, stimulation of B and /or T cells, and stimulation of macrophages.
  • An "immunogenic response" to an antigen or composition is the development in a subject of a humoral and/or a cellular immune response to an antigen present in the composition of interest.
  • a “humoral immune response” refers to an immune response mediated by antibody molecules
  • a “cellular immune response” is one mediated by T-lymphocytes and/or other white blood ceils.
  • CTL cytolytic T- cells
  • CTLs have specificity for peptide antigens that are presented in association with proteins encoded by the major histocompatibility complex (MHC) and expressed on the surfaces of cells.
  • helper T-cells help induce and promote the destruction of intracellular microbes, or the lysis of cells infected with such microbes.
  • Another aspect of cellular immuni ty involves an antigen- specific response by helper T-cells.
  • Helper T-cells act to help stimulate the function, and focus the activity of, nonspecific effector cells against cells displaying peptide antigens in association with MHC molecules on their surface.
  • a "cellular immune response” also refers to the production of cytokines, chemokines and other such molecules produced by activated T-cells and/or other white blood cells, including those derived from CD4+ and CD8+ T-cells.
  • an immunological response may include one or more of the following effects: the production of an tibodies by B-celis; and/or the acti vation of suppressor T-celis and/or ⁇ T-cells directed specifically to an antigen or antigens present in the composition or vaccine of interest.
  • These responses ma serve to neutralize infeetivity, and/or mediate antibody-complement, or antibody dependent cell cytotoxicity (ADCC) to provide protection to an immunized host.
  • ADCC antibody dependent cell cytotoxicity
  • Such responses can be determined using standard immunoassays and neutralization assays, w r ell known in the art.
  • the term "host” refers to humans as well as other animals.
  • mucosal delivery relates to delivery of a composition to a mucous membrane, such as the mucosa of the gastrointestinal tract (e.g., the buccal or labial mucosa) or the mucosa of the respiratory tract (e.g. , the nasal mucosa).
  • a mucous membrane such as the mucosa of the gastrointestinal tract (e.g., the buccal or labial mucosa) or the mucosa of the respiratory tract (e.g. , the nasal mucosa).
  • One aspect of the invention relates to methods for production and purification of capsid proteins and VLPs derived therefrom (See, Expression and self-assembly of empty virus-like particles of hepatitis E virus.
  • Li TC Yamakawa Y, S uzuki K, Tatsumi M, Razak MA, Uchida T, Takeda N, Miyamura T., J Virol. 1997 Oct;71(10):7207-13.
  • Li TC Takeda N, Miyamura T, Matsuura Y, Wang JC, Engvail H, Hammar L, Xing L, Cheng RH. J Virol 2005 Oct;79(20): 12999-3006.
  • Niikura M et al Chimeric recombinant hepatitis E virus-like particles as an oral vaccine vehicle presenting foreign epitopes.
  • the capsid proteins are modified capsid proteins and the VLPs derived therefrom are cysteine modified HEV VLPs.
  • the modified capsid proteins contain one or more cysteine residues in a surface variable loop of HEV OR 1 2. or a portion thereof.
  • Various expression systems can be used to express the capsid proteins of the present invention.
  • Examples of expression systems useful for the production of vims- like particles of the present invention include, but are not limited to, bacterial expression system (e.g.,
  • Preferred expression system of the present invention includes baculovirus expression systems using insect cells.
  • General methods for example, for handling and preparing baculovirus vectors and baculoviral DNA, as well as insect cell cul ture procedures, are outlined in A Manual of Methods for
  • capsid proteins of the present invention can be cloned into the baculovirus vector, and used to infect appropriate host cells (see, for example, O'Reilly et al,
  • An insect cell line ⁇ e.g., Sf9 or Tn5
  • a transfer vector containing polynucleic acids which encodes the capsid proteins of the in venti on.
  • Th e tran sfer vector in cludes, for example, linearized baculovirus DNA and a plasmid containing the desired polynucleotides.
  • the host cell line may be co-transfected with the linearized baculovirus DNA and a plasmid in order to make a recombinant baculovirus.
  • the modified capsid proteins or VLPs derived therefrom can be chemically conjugated to one or more bioactive agents.
  • one or more cysteine residues of the capsid proteins can be acyiated, alkylated, aryiated, succinyiated, or oxidized using methods known in the art.
  • the one or more cysteine residues can be conjugated using a maleimide functional group to covalentiy conjugate a bioactive agent to the thiol moiety of the cysteine, in some cases, the bioactive agent can be modified to introduce a maleimide functional group using CLICK chemistry.
  • an alkyne deri vative of the bioactive agent can be contacted with a maieimide-azide in the presence of C11SO4 and ascorbic acid to produce a maleimide bioactive agent.
  • the maleimide can then be contacted with the one or more cysteines of the modified capsid protein to covalentiy link the two molecules.
  • the conjugating is performed on capsid protein that is not assembled into a VLP (e.g. , in the presence of EDTA, EGTA, and/or a reducing agent such as DTT or betamercaptoetha.no i).
  • the conjugating is performed on capsid protein that is assembled into a VLP.
  • HEV virus-like particles e.g., cysteine modified HEV VLPs
  • Any standard technique in the art can be used to encapsulate a heterologous nucleic acid, protein, polypeptide, chemotherapeutic, imaging agent, nanoparticle, etc.
  • the general proceed ure involves (1) disassembling the VLPs formed by the capsid protein according to the present invention; and (2) reconstructing the VLPs in the presence of the bioactive agent.
  • a skilled artisan would recognize that it is preferred to have purified VLPs before the encapsulation procedure. It is particularly preferred to have the VLPs depleted of, or substantially depleted of, any undesired materials (e.g., nucleic acids) before the encapsulation procedure.
  • Disassembly of VLPs can be carried out using any standard technique in the art. Reconstituted virus-like particle can be produced under physiological conditions (See, US Patent Publication No.: 20080131928). Often, disassembly of virus-like particles requires an agent to disrupt the assembly of VLPs, such as a reducing agent or a chelating agent (See, US Patent Publication No.: 20040152181). A skilled artisan would recognize that factors and conditions that affect assembly and disassembiy include: pH, ionic strength, posttranslational modifications of viral capsid proteins, disulfide bonds, and divalent cation bonding, among others.
  • the VLPs of the present invention is disassembled by the removal of calcium ions (See, Touze A, Coursaget P. in vitro gene transfer using human papillomavirus- like particles. Nucleic Acids Res 1998; 26: 1317-1323; Takamura et a!., DNA vaccine- encapsulated virus-like particles derived from an orally transmissible virus stimulate mucosal and systemic immune responses by oral administration. Gene Therapy 2004; 1 1 :628-635).
  • a reducing agent or a chelating agent or both are used to disassemble the VLPs.
  • Various reducing agents can be used.
  • Preferred embodiments of the reducing agents include, but are not limited to, dithiothreitol (DTT).
  • DTT dithiothreitol
  • Various chelating agents can be used, e.g., ethylene glycol tetraacetic acid (EGTA) or ethyl enediarninetetraacetic acid (EDTA).
  • Examples of VLP disassembly conditions include, but are not limited to, the following: purified VLPs were disrupted by incubation of a buffer containing 50 mM Tris- HC1 (pH 7.5), 150 mM NaCf, 1 mM EGTA and 20 mM dithiothreitol for 30 minutes.
  • VLPs complete disassembly of the VLPs is not required, although preferred, to encapsulate a bioaciive agent.
  • An artisan would also recognize that, on other occasions, it is preferred to have partial disassembly of the VLPs.
  • the conditions for the partial disassembly of the VLPs can be controlled to still allow efficient encapsulation of a bioaciive agent.
  • Partial disassembly of the VLPs can be achieved by treatment of VLPs with reducing agents alone (e.g., 20 mM DTT) (Sapp et al, j . Gen. Virol., 76:2407-2412, 1995.).
  • encapsulation of a bioactive agent can be carried out by reassembling the VLPs in the presence of the bioactive agent.
  • a bioactive agent having a net negative charge it can be advantageous to utilize a bioactive agent having a net negative charge to enhance encapsulation.
  • nucleic acids have a net negative charge and can be preferentially encapsulated as compared to compounds that have a positive or neutral charge.
  • reassembly of the VLPs is achieved by re-supplementation of calcium ions to the disrupted V LPs.
  • reassembly of the V LPs is achieved by removal of the reducing agents or the chelating agents.
  • factors such as pH and ionic strength, other factors described in the present inven tion, can be adjusted to achieve effi cient reassembly of the VLPs and efficien t encapsul ation of the bioactive agent.
  • encapsulation is performed as follows: Following 30 mm of incubation at room temperature, a bioactive agent in 50 mM Tris-HCl buffer (pH 7.5) and 150 mM aCl is added to the disrupted VLP preparation . The disrupted VLP preparation is then refolded by incubation for 1 h with increasing concentrations of Ca(3 ⁇ 4 up to a final concentration of 5 mM. VLPs are pel leted by ultracentrifugation and resuspended in 10 mM potassium-MES buffer (pH 6.2).
  • VLPs are purified from any unencapsulated bioactive agent and disrupted with EGTA (1 mM). Absorbance of the supernatant, or other suitable methods can be used for detection of the bioactive agent,
  • the bioactive agent or imaging agent to be encapsulated is conjugated to an encapsidation signal.
  • an RNA element corresponding to codons 35-59 of HEV open reading frame 1 is a powerful encapsidation signal, allowing specific interaction in vitro with HEV capsid protein, including truncated and/or cysteine modified versions of HEV ORF2 VLP as described herein.
  • VLP as a carri er for therapeutic or imaging agents
  • chemical linkers e.g., LC-SPDP or aptamer, teiodendrimers
  • tag the agent ⁇ e.g., chemotherapeutic with an HEV encapsidation signal like the foregoing RNA element can be used prior to the capsid self-assembly.
  • a detectable label (imaging agent) is encapsulated.
  • the detectable label can be a moiety renders a molecule to which it is attached to detectable by a variety of mechanisms including chemical, enzymatic, immunological, or radiological means.
  • detectable labels include fluorescent molecules (such as fluorescein, rhodamine, Texas Red, and phycoerythrin) and enzyme molecules (such as horseradish peroxidase, alkaline phosphatase, and ⁇ galactosidase) that allow detection based on fluorescence emission or a product of a chemical reaction catalyzed by the enzyme.
  • Radioactive labels involving various isotopes can also be attached to appropriate molecules to enable detection by any suitable methods that registers radioactivity, such as autoradiography. See, e.g., Tijssen, "Practice and Theory of Enzyme immunoassays," Laboratory Techniques in Biochemistry and Molecular Biology, Burdon and van Knippenberg Eds., Elsevier (1985), pp. 9 20.
  • a bioactive agent is encapsulated.
  • the bioactive agent is a chemotherapeutic.
  • Suitable chemotherapeutics include, but are not limi ted to, cytotoxic drugs.
  • cytotoxic drugs whi ch may be used in the present invention include: alkylating drugs, such as cyclophosphamide, ifospfamide, chlorambucil, meiphalan, busulfan, lomustine, carmustine, chlormethhine (mustine), estramustine, treosulfan, thiotepa, mitobronitol; cytotoxic antibiotics, such as doxorubicin, epirabicin, aciarubicin, idarubicin, daunorubicin, mitoxantrone (mitozantrone), bleomycin, dactinomycin and mitomycin; antimetabolites, such as methotrexate, capecitabine; cytarabine, fludarabine, cladribine, gemcitabine, fluorouracil, raltitrexed (tomudex
  • one or more of the foregoing imaging agents and/or bioactive agents, or a combination thereof can additionally or alternatively be conjugated to a cysteine (e.g., recombinantly introduced cysteine) in a P-domain surface variable loop or C-terminus vi a a thiol, linkage.
  • a cysteine e.g., recombinantly introduced cysteine
  • one or more of the foregoing imaging agents and/or bioactive agents, or a combination thereof can additionally or alternatively be conjugated to a second cysteine ⁇ e.g. , recombinantly introduced cysteine) in a P-domain surface variable loop or C-terminus via a thiol linkage.
  • the size of the VLPs can vary when different constructs of the capsid protein are used.
  • the N-tenninal portion of the capsid protein can be adjusted to increase or decrease the size and encapsulation capacity of the VLPs.
  • a portion of HEV ORF 3 protein fused to the N- terminal of a portion of HEV ORF 2 proteins is utilized to adjust the size of the V LPs.
  • the HEV VLP is formed from a portion of HEV ORF2 having at least residues 1 12-608 of HEV ORF 2.
  • the present invention also provides pharmaceutical compositions or physiological compositions comprising a cysteine modified HEV VLP formed by the capsid protein of the present invention.
  • Such pharmaceutical or physiological compositions also include one or more pharmaceutically or physiologically acceptable excipients or carriers.
  • Pharmaceutical compositions of the invention are suitable for use in a variety of drug delivery systems.
  • Suitable formulations for use in the present invention are found in Remington 's
  • compositions of the present invention can be administered to a host with an excipient.
  • Excipients useful for the present invention include, but are not limited to, vehicles, binders, disintegrants, fillers (diluents), lubricants, giidants (flow enhancers), compression aids, colors, sweeteners, preservatives, suspending dispersing agents, film formers/coatings, flavors and printing inks.
  • adjuvants can be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacender Calmette Guerin) and Corynebacterium parvum. Such adjuvants are also well known in the art. Further adjuvants that can be administered with the compositions of the invention include, but are not limited to, Monophosphoryl. lipid immunomodulator, AdjuVax 100a, QS 21 , QS 18,
  • compositions and methods of the present invention can be administered to stimulate immune response without an adjuvant. Therefore, the compositions of the present invention are in some cases administered to a host without an adjuvant.
  • compositions of the present invention are suitable for oral delivery. Because mucosal surfaces are inter-connected, stimulation of one mucosal surface by an antigen can induce mucosal immunity not only on the directly stimulated surface, but also on the distant ones.
  • oral delivery of the compositions of the present invention can protect against respiratory and genital -urinary infections.
  • the compositions of the present invention are also suitable for mucosal delivery, such as delivery to the buccal or labial mucosa or the respiratory tract mucosa, including the nasal mucosa.
  • the pharmaceutical composi tions of the present invention can be administered by various routes, e.g.
  • oral subcutaneous, transdermal, intradermal, intramuscular, intravenous, or intraperitoneal .
  • the preferred routes of administering the pharmaceutical compositions are oral deliver ⁇ ' at daily doses of about 0.01-5000 nig, preferably 5-500 mg.
  • the appropriate dose may be administered in a single daily dose or as divided doses presented at appropriate intervals, for example as two, three, four, or more subdoses per day.
  • inert and pharmaceutical ly acceptable carriers are used.
  • the pharmaceutical carrier can be either solid or liquid.
  • Solid form preparations include, for example, powders, tablets, dispersible granules, capsules, cachets, and suppositories.
  • a solid carrier can be one or more substances that can also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material .
  • the carrier is generally a finely divided solid that is in a mixture with the finely divided active component, e.g., a chimeric virus-like particles with an encapsulated nucleic acid.
  • the active ingredient a chimeric virus-like particles with an encapsulated nucleic acid
  • the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
  • a low- melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient-sized molds and allowed to cool and solidify.
  • Powders and tablets preferably contain between about 5% to about 70% by weight of the active ingredient.
  • Suitable carriers include, for example, magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low-melting wax, cocoa butter, and the like.
  • compositions can include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component (with or without other carriers) is surrounded by the carrier, such that the carrier is thus in association with the compound, in a similar manner, cachets can also be included. Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration.
  • Liquid pharmaceutical compositions include, for example, solutions suitable for oral or parenteral administration, suspensions, and emulsions suitable for oral administration.
  • Sterile water solutions of the active component e.g., a chimeric virus-like particles with ars encapsulated nuc leic acid
  • solvents comprising water, buffered water, saline, PBS, ethanol, or propylene glycol
  • the compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents, and the like.
  • Sterile solutions can be prepared by suspending the active component (e.g., a chimeric virus-like particles with an encapsulated nucleic acid) in the desired solvent system, and then passing the resulting solution through a membrane filter to sterilize it or, alternatively, by dissolving the sterile compound in a previously sterilized solvent under sterile conditions.
  • the resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration.
  • the pH of the preparations typically will be between 3 and 9, more preferably from 5 to 8, and most preferably from 6 to 7.
  • compositions of the present invention can be administered for prophylactic and/or therapeutic treatments.
  • compositions are administered to a patient already suffering from a condition in an amount sufficient to prevent, cure, reverse, or at least partially slow or arrest the symptoms of the condition and its complications.
  • An amount adequate to accomplish this is defined as a "therapeutically effective dose.” Amounts effective for this use will depend on the severity of the disease or condition and the weight and general state of the patient, but generally range from about 0.1 mg to about 2,000 mg of the composition per day for a 70 kg patient, with dosages of from about 5 mg to about 500 mg of the composition per day for a 70 kg patient being more commonly used.
  • compositions of the present invention are administered to a patient susceptible to or otherwise at risk of developing a disease or condition, in an amount sufficient to delay or prevent the onset of the symptoms.
  • an amount is defined to be a "prophylactically effective dose.”
  • the precise amounts of the composition again depend on the patient's state of health and weight, but generally range from about 0.1 mg to about 2,000 mg of the inhibitor for a 70 kg patient per day, more commonly from about 5 mg to about 500 mg for a 70 kg patient per day,
  • compositions can be carried out with dose levels and pattern being selected by the treating physician.
  • pharmaceutical formulations should provide a quantity of composition of the present invention sufficient to effectively stimulate immune response in the patient, either therapeutically or prophylatically.
  • VLPs Virus-like particles
  • This application relies mainly on the property of self-assembly as well as the ease of genetic modifications, to fulfill the designed application for the given VLP.
  • chemical conjugation of foreign peptides to VLP displays a significant advantage because it al lows a great variety of entities, such as peptide or oligosaccharide, to be conjugated to the surface of vims like particles in modulated and flexible manner without alteration of VLP assembly.
  • Hepatitis E virus is composed of a non-enveloped icosahedral capsid, enclosing a single stranded RNA genome of 7.2 kilo bases.
  • the major capsid protein is encoded by the second open reading frame (ORF2) and is essential not only for virus assembly, but also for immunogenicity and host interaction. 6
  • the recombinant capsid protein (PORF2) is able to self-assemble into VLPs when expressed in insect cells after deletion of 111 amino acids from the N -terminal end and 52 amino acids from the C-terminal end.
  • PORF2 folds into three domains: S (shell; amino acids 1 18-317), M (middle; amino acids 318-451) and P (protruding; amino acids 452-606).
  • ⁇ 11 Recombinant HEV-VLPs consists of 60 copies of PORF2, in which the S-domain possesses typical eight anti-parallel folding and stabilizes icosahedral shell and the P-domain protrudes as a surface spike that contributes greatly to HEV antigenicity.
  • HEV-VLP can be readily manipulated to, e.g., alter antigenicity, reduce the size from a T3 icosahedral particle to a Tl icosahedral particle, and/or modify the sequence at the P-domain without interfering with HEV-VLP assembly.
  • HEV- VLP can be stable in acidic environment' ' and resistant to proteolytic digestion, 13 thus it poses a great advantage as an oral delivery vehicle.
  • oral administration of HEV- VLP elicits both systemic and mucosal immunity without eliciting tolerance, and thus can provide protective immunity against HEV challenge in non- human primates 15
  • Chimeric VLPs carrying a foreign epitope are able to elicit mucosal and systemic antibodies against both HEV and the foreign epitope after oral administration.
  • HEV-VLP can orally deliver plasmid DNA to the epithelial cells of the small intestine and induce antibody and cytotoxic T lymphocyte (CTL) responses against the plasmid encoded antigen.
  • CTL cytotoxic T lymphocyte
  • the modified HEV-VLP after conjugation with LXY30, a ligand peptide with a high affinity for human malignant breast tumor cells, 58 showed specific targeting to breast tumor ceil both in vitro and in vivo, indicating that delivery route of viruslike particle can be manipulated to facilitate targeted delivery of chemotberapeutic drugs or siRNA to pathologic foci.
  • Aikyiation and acylation are the two traditional bioconjugation methods that are used to covendedly modify virus particles by attaching a ligand at amine group of lysine or thiol group on cysteine, respectively.
  • HEV ORF2 naturally has four lysine residues that are
  • the P-domain carries the antigenic structure of HEV
  • sequence mutation may alter the immunoreactivity of HEV antibodies against the obtained chimeric VLPs.
  • the antigenicity of the chimeric N573C VLP to the wild type HEV- VLP in the presence or absence of biotin conjugation was compared.
  • An ELISA plate was coated with N573C VLP without conjugation, maleimide-biotin conjugated N573C VLP, or the wild type HEV-VLP and then incubated with two monoclonal antibodies, HEP230 or HEP40-4.
  • HEP230 In contrast to HEP40-4, HEP230 exhibited strong binding for the wild type HEV-VLP, but failed to exhibit detectable binding to N573C VLP regardless of biotin conjugation (Fig 2B). The ELISA against the mixture of HEP230 and HEP40-4 showed similar trend (data not shown). This data suggested that residue N573 is important to HEP230 binding. Residue N573 is Involved in FA.B230 Binding
  • connection density between the spikes was much weaker than the capsid and was not big enough to accommodate the Fab molecule.
  • the size of the connection density appeared to be in good agreement with the contacting surface of the Fab molecule (Fig 3B), indicating that the Fab230 may contact VLP at this position.
  • LXY3Q Conjugated to N573C VLP Shows Cancer Cell Targeting [ ⁇ 090]
  • a breast cancer cell targeting ligand L.XY30
  • L.XY30-VLPs The LXY30-decorated N573C-VLPs (LXY30-VLPs) remained intact after the chemical conjugation (Fig. 4).
  • Flow cytometry revealed a significantly higher attachment (>5 fold) for LXY30-VLPs to MDA-MB-231 breast cancer cells, compared to that for the wild type VLPs (Fig.
  • LXY30-VLPs enter the MDA-MB-231 cells.
  • the LXY30-VLPs were labeled with the near infrared dye, Cy5.5, and incubated with MDA-MB-231 cells at 37 °C for 1 hour.
  • Fig 5B most Cy5.5-labeled VLP distributed around the perinuclear region of MDA-MB-231 cells, which meant the VLP was internalized into the cytoplasm.
  • LXY30 and scrambled LXY30 was also tested for uptake by the glioma cell line U-87MG, suggesting its use as a conjugate for HEV -VLP for targeting to glioma ceils (Fig. 6).
  • Noninvasive in vivo imaging has been shown to be a powerful tool to visually monitor the delivery of therapeutic reagents.
  • Both Cy5.5 labeled LXY30-VLPs and the Cy5.5 labeled wild type VLPs were intravenously injected via tail vein in nude mice bearing DA-MB-23.1 xenografts, respectively. The mice were scanned with the fluorescence imager at various intervals for up to 72 hours.
  • HEV as an enteric transmitted virus, preferentially enters hepatocytes in liver, it is not surprising that Cy5.5 labeled VLPs accumulated within one hour at the abdominal organs including liver. Ex vi vo imaging of excised organs were performed at 72 hours post injection. The remaining signal of Cy5.5 was found predominantly at the liver and kidney, however, the signal appeared weak in excised tumor (data not shown). The high uptake in liver and kidney could be explained in part as leaking of the breakdown VLPs from circulating vessels. Overall, these results indicate that Cy5.5/LXY30 linked HEV- VLPs is able to target MDA-MB-231 xenografts in vivo.
  • VLPs As a drug delivery system for diseases that require the delivery of cytotoxic drugs or specific genetic modifications, ceil tagging, or drugs that require encapsulation .
  • VLPs provide an opportunity for tissue/cell specific approaches while minimizing the overall damage to healthy cells.
  • HEV-VLP like the Hepatitis E vims can maintain its structure through the low pH and proteolytic
  • HE V-VLPs could have a higher propensity for efficient delivery and reduced
  • the streptavidin homotetramer can intercalate into the surface depression and form multivalent interaction with N573C anchored biotin molecules, which largely reduces the rate of dissociation.
  • the avidity of streptavidin to N573C VLP is the highest although the affinity of streptavidin to a single biotin appeared to be the same as it showed by its binding to the disassembled VLPs.
  • the quaternary arrangement of cysteine residues in the other chimeric VLPs does not support multivalent binding of streptavidin.
  • N573 residue is located within the binding footprint of Fab (Fig 3), so that mutation of N573 blocks the interaction of PORF2 and HEP2.30.
  • the volume of a fivefold depression is not sufficient to s multaneously accommodate five Fab molecules, so that the structure of Fab-bound VLP can only capture the conta cting region of the Fab mo lecule after icosahedra!-average.
  • the density is about 20% occupancy to that of the capsid, suggesting that only one Fab molecule resided at each five-fold axis. This is consistent with the non-icosahedral symmetry averaged density map.
  • HEV-573C VLPs were chosen as a platform of modularized theranostic capsule because of its possibility of weakening the immunoresistance against the pre-existing HEV antibodies as well as the possibility of multi valent binding with the ligand.
  • thiol-selective conjugation for coupling a cancer cell targeting ligand to VLP, because of the spatial specificity provided by cysteine replacement.
  • the conjugation of dye was performed by amine-selective conjugation that gave rise to sufficient MR signal for us to detect VLP distribution in MDA-MB-231 breast cancer ceils in vitro and in vivo (in mice). Therefore, the HEV-Cys VLPs can be used as a platform for dual-functional, tagging with cancer adliesion-ligand and a detection marker, a robust candidate for cancer diagnosis.
  • HEV-Cys- VLP is capable of encapsulation of a variety of bioactive substances.
  • HEV-Cys- VLPs can encapsulate magnetic nano-particles such as ferrite, for both diagnosis under MR! and thermotherapy. in such cases, the magnetic nano- particies can be selectively stimulated using ultrasound or radio frequenc electromagnetic radiation to heat the ferrite particles encapsulated in the tumor targeted theroanostic capsule.
  • Negatively charged micro-RNA/siRNA could be also encapsulated into the interior of HEV-Cys-VLPs. ' By binding the cancer cell targeting ligand on its surface, HEV-Cys-VLP could have potential applications in cancer cell targeting gene therapy;
  • baculovirus-based expression vectors The recombinant baculoviruses used to express the mutated HEV ORF2 proteins were generated using the Bac-to-Bac® Baculovirus Expression System (In vitro gen) on Sf9 cells (Invitrogen) and each of the pFastBacl/ HE V ORF2 plasmids described above according to the protocols supplied by the manufacturer.
  • the Sf9 and High Five cells were maintained on ESF921 medium (Expression Systems, Davis, CA USA) and Exceli 420 medium (SAFC Biosciences, Lenexa, KS) supplemented with 2.5% heat-inactivated FBS, respectively, following standard protocols. 24
  • ESF921 medium Expression Systems, Davis, CA USA
  • Exceli 420 medium SAFC Biosciences, Lenexa, KS
  • FBS heat-inactivated FBS
  • VLPs were collected and purified through multiple steps of CsCl equilibrium densit gradient ltracentrifugation as described previously.' The purified VLPs were resuspersded in 10 mM potassium-MES buffer, pH6.2, and stored at 4 °C. A Mini dialysis device (Miliipore) was used in the disassembly experiments. The purified VLPs were disrupted by dialysis against buffer containing EDTA (lOmM) or/and DTT (20mM).
  • the purified VLPs were loaded onto a glow-discharged, carbon -coated EM grid and stained with 2% uranyl acetate and examined under a JOEL. JEM-1230 transmission electron microscope at a magnification of 3Q.00QX. The images were recorded on a CCD camera (TVIPS Gauting, Germany).
  • PVDF polyvinyl! dene fluoride
  • HRP conjugated streptavidin at a ratio of 1 : 10,000.
  • the biotin- streptavidin reaction was then detected by an enhanced chemiluminescence method using an ECL kit (Amersham Biosciences, Piscataway, NJ).
  • Both recombinant HEV -WT-VLP particles including HEV-WT and HEV-Cys mutants were prepared in 10 mM potassium-MES, pH 6.2 coating buffer.
  • the proteins were diluted to the final concentrations of 1-100 ng/ml and coated overnight at 4 °C onto a clear bottom 96-well plate (Nunc, Pleasant Prairie, WI). Control wells were inclubated with 0.01 M PBS, pH 7.2.
  • the coated VLPs were incubated with ⁇ 50 ⁇ of the HEV antibodies for two hours at 37 °C after blocking with 0.5% Tween-20 in Tris buffer containing 20 mM Tris, pH 7.4, and 150 mM NaCl.
  • the associated HEV antibodies were detected using alkaline phosphatase-labeled anti-mouse FAb.
  • the enzymatic reaction was developed using pnitrophenyiphosphate (pNPP) solution (Sigma Co).
  • pNPP pnitrophenyiphosphate
  • the yellow color product of nitrophenyi was measured at 405 ra using a microplate reader and the average absorbance value of each VLP was calculated.
  • Maleirnide-conjugated LXY30 was prepared by mixing 650 ⁇ maleimide-azide and 650 ⁇ aikyne-LXY ⁇ 3Q in the presence of 200 ⁇ CuS04 and 1 mM ascorbic acid at 4 °C overnight. Then maleirnide-conjugated LXY30 was added at molar ratio of 3: 1 (ligand vs. binding site) to react with the C s residues on HEV-Cys mutants though a thiol reaction at 4 °C overnight. The unconjugated molecules were removed by a 7,000 MWCO desalting columns, (ZebaTM Spin Desalting Columns, Thermo Scientific).
  • Cy5.5 NHS ester (200nm) was used to react with HEV-Cys mutants (20 ⁇ ) at molar ratio of 300: 1 (dye vs VLP) in buffer containing 0.01 M PBS, pH 7.2 at room temp for 2 hrs, following by incubation at 4 °C overnight.
  • the free Cy5.5 NHS ester was then removed by a 7,000 MWCO desalting column, (ZebaTM Spin Desalting Columns, Thermo Scientific). Flow Cytometry
  • MDA-MB-231 breast cancer cells were incubated with Cy5.5-labeled HEV-573C VLPs (VLP-Cy5.5) or Cy5.5/LXY30 conjugated HEV-573C VLPs (LXY3-VLP Cy5.5) for 1 hour at 37 °C, respectively. Then the cells were washed with PBS 3 times and re-suspended in PBS for the flow cytometric analysis. 1 A total of 0, 000 events were collected for each sample. Unstained ceils were used as a control.
  • mice Female SPF BALB/c mice, 10-12 weeks age, were purchased from Charles River (Davis, CA)) and kept under pathogen-free conditions according to AAALAC guidelines and were allowed to acclimatize for at least 4 days prior to any experiments.
  • DA-MB-231 breast cancer cells were injected subcutaneously into nude mice to form subcutaneous nodules. All animal experiments were performed in compliance with institutional guidelines and according to protocol No. 06-12262 approved by the Animal Use and Care
  • mice were intravenously injected through the tail vein with 4 nmol/L Cy5.5 fluorescent-labeled HEV-573C VLPs (VLP-Cy5.5) and LXY30 conjugated HEV-573C VLPs (LXY30- VLP-Cy5.5) respectively.
  • mice were scanned with Kodak imaging system IS20QQMM according to the procedure described previously." Images were collected with an excitation bandpass filter at 625 nm and an emission at 700 nm, exposure time was 30 s per image. After in vivo imaging, animals were euthanized by C0 2 overdose at 72 h after injection. Tumors, organs, and muscle tissue were excised and imaged with the Kodak imaging station. Preparation of VLP-Fab Complexes for Cryoelectron Microscopy
  • VLP-Fab complexes were prepared by incubating VLPs and Fabs at a molar ratio of 1 : 180 at 4 °C overnight. To ensure optimal purity, maxima] occupancy, and ultimately reduced background noise during structural determination, pure VLP-FAB complexes were obtained by passing the sample through a short Sephacryl-300 gel -filtration column. Optical density readings at a wavelength of 280 nm were used to select the fractions that contained VLP-Fab complexes. The purified VLP fraction was then characterized by SDS-PAGE under reducing condition to validate the binding of Fab.
  • Figure 10 depicts various routes for modifying the surface of HEV ORF2 VLP nanocarriers and encapsulation of detectable labels, drugs, or nucleic acid payloads.
  • An HEV ORF2 VLP-based nanocarrier can self-assemble in vitro and encapsulate anticancer therapeutics or image reagents. With genetic engineering, cancer targeting iigands can be conveniently ligated to the surface of the carrier for cancer-specific targeted delivery.
  • the chimeric VLP exhibits excellent biocompacibility, high absorption of near- infrared light, and structure-dependent fluorescence quenching/emission.
  • This nanomedicme platform allows integration of multiple imaging and therapeutic modalities for superior cancer detection and therapy.
  • Intratumoral delivery is a challenge task for modem medical research and is a process that comprises lots of negotiations betw een nanocarriers with cells, extracellular matrix, and the vasculature inside the tumor.
  • Described herein is an HEV -VLP based nanocarrier for targeted tumor delivery guided by imaging the in situ distribution of VLP at the whole animal level, to the tumor interior, and ultimately down to subcellular organelles.
  • Engineered HEV-VLPs for tumor targeting are provided by adding adhesion tag(s) to the surface of the P-domain.
  • a genetical ly inserted a fluorescence marker can also be inserted into the VLP so that it can be used as endogenous reporter.
  • Use of these nanoparticles demonstrate the power of correlative microscopic studies on the role it plays in the development of nanocarriers.
  • Nanocarriers composed of synthetic polymers, liposomes, and dendrimers, have been well-investigated, however, there are only two FDA-approved antibody-conjugates and four FDA-approved nanoparticle-based drug delivery carriers.
  • Virus like particle is natural provided nanocarriers that possess several attractive features. They can be self-assembled into monodisperse, nansized particles, with defined periodicity that enables dense, repetitive display of tumor targeting agents. In addition, the V LP possesses multiple unique features that make it more attractive as potential nanocarrier.
  • HEV VLP nanoparticles can be characterized with electron microscopy in the presence or absence of contrast enhancing reagents. Microscopic characterization of nanoparticles can be routinely performed to examine the physical properties of the nanocarriers using methods described herein, such as imaging frozen hydrated
  • multifunctional particles e.g. , using contrast enhanced gold atom clusters (Fig. 1 1A).
  • Methods and compositions for conjugating gold clusters to cysteine thioiates include direct ligand exchange and/or maleimide cysteine conjugation (see, e.g., Ackerson ei al, Bioconjug. Chem., 21 , 214-218 (2010); Ackerson et al., Methods EnzymoL, 481 : 195-230 (2010); and Maijormaki et al, PNAS, 11 1(4): 1277 ⁇ 81 (2014)).
  • the direct visualization by electron microscopy precisely reveals the size, shape, and domain arrangement of the nanoparticles, as well as the protein/drug absorption on nanoparticles, factors that can influence the overall Inactivity of the nanoparticles and the efficacy of targeted tumor delivery .
  • the direct visualization by electron microscopy precisely reveals the size, shape, and domain arrangement of the nanoparticles, as well as the protein/drug absorption on nanoparticles, factors that can influence the overall Inactivity of the nanoparticles and the efficacy
  • nanoparticles can be imaged with either by cryo-electron microscopy on unstained frozen hydrated specimen or by traditional electron microscopy on negatively stained specimen.
  • the cryoEM is well-suited to imagine the biological macromolecular complexes (Fig. 11 A- D).
  • correlative microscopic techniques ca be based on these newl described conjugates to achieve the utilization of multiple imaging modalities in the examination of the same tumor specimen in order to deliver information above and beyond the capability of either modality alone.
  • li ght and electron microscopy can be used to image from the organismal level, to the organ level, to cells, and even subcellular details.
  • MR Near-Infrared
  • CLEM correlative light and electron microscopy
  • VLP intratumorai distribution With, e.g., high pressure freezing and freeze substitution, tumor diagnosis can be preserved to maintain the sufficient antigenicity of HEV-VLP for immunofluorescence labeling.
  • Region identification is one of the critical issues for tumor diagnosis, where both light and electron microscopies indeed image the same object.
  • the coordinates of 1 ) photoconversion of DAB (3,3 '-diammobenzidine tetrahydrochrloride); 2) dual functional probe, FluoroNanogold; and 3) EM finder grids can be mformative in multiple modalities of tumor identification.
  • the first method uses free oxygen radicals that formed upon illumination of fluorochromes to induce oxidation of DAB in situ.
  • FluoroNanogold conjugated antibody is a commercially available reagent that provides both light and electron microscope contrast by conjugation of 1 ,4 nm gold clusters either with green or with red fluorophore. Unlike colloidal gold of other sizes, the 1.4 nm nanogold cluster does not quench fluorescence.
  • HEV VLPs described herein allow detection and visualization by 3D electron tomography to monitoring drug delivery.
  • Electron tomography provides 3D spatial information to describe the biodistribution of drug delivery vehicles inside the tumor. In a three-dimensional entity, the microenvironment of tumor can be much more complex than what found in vitro with cultured monolayer cells. Electron tomography can be used as a complement to C LEM in analyzing the intratumoral accumulation of drug- loaded V LP, so as to elucidate the mechanism for VLP extravasation and passing through the cell layers inside a tumor.
  • HEV VLPs can be equipped and encapsulated with additional imaging agents, e.g., water-soluble CdSe/ZnS quantum dots or Ferrite, in a disassembly-and-reassembly process (Fig 12).
  • additional imaging agents e.g., water-soluble CdSe/ZnS quantum dots or Ferrite
  • Quantum dots can provide an dual-functional probe to track VLP tumor uptake with correlative light and electron microscopy.
  • the efficiency of packaging can be optimized to have efficient encapsulation, e.g., by coating the quantum dots with an HEV encapsidation signal through LC-SPDP- mediated crosslinking.
  • RNA element occupying codons 35-59 of HEV open reading frame 1 is a powerful encapsidation signal, allowing specific interaction in vitro with HEV capsid protein, including truncated and/or cysteine modi fied versions of HEV ORF2 VLP as described herein.
  • VLP as drag carrier, chemical linkers (e.g., LC-SPDP or aptamer, telodendrimers) that tag the drug (e.g., chemotherapeutic) with an HEV encapsidation signal like the foregoing RNA element can be used prior to the capsid self-assembly.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Virology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Physics & Mathematics (AREA)
  • Nanotechnology (AREA)
  • Optics & Photonics (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

Modified capsid proteins containing at least a portion of hepatitis E virus (HEV) open reading frame 2 (ORF2) having one or more cysteine residues in a surface variable loop or the C-terminus of HEV ORF2, or a portion thereof, are provided. The modified capsid proteins can be used to form hepatitis E virus (HEV) virus like particles (VLPs) having cysteine functional groups exposed on the outer-surface. The exposed cysteine functional groups can be modified via their thiol reactive group. For example, a bioactive agent, such as a cell-targeting ligand, can be conjugated to the one or more cysteines for targeted delivery of chemically activated nanocapsids.

Description

CHEMICALLY ACTIVATED NANOCAPSID FUNCTIONALIZED FOR
CROSS-REFERENCE TO RELATED APPLICATION
[Θ001] This application claims prioity to U.S. Provisional Application No. 62/000,465, filed on May 19, 2014, the contents of which are hereby incorporated by reference in the entirety for ail purposes.
STATEMENT AS TO RIGHTS TO INVENTIONS MADE UNDER
FEDERALLY SPONSORED RESEARCH AND DEVELOPMENT
[0002] This inventio was made with government support under contract AI095382 awarded by the National Institutes of Health. The government has certain rights in the invention.
BACKGROUND OF THE INVENTION
[0003] Vims-like particles (VLPs) can serve as nanocarriers for targeted delivery of diagnostics and therapeutics regimes, such as DNA/RNA and a variety of chemotherapeu ics. Hepatitis E virus (HEV) is an enteric-transmitted virus that causes acute liver inflammation in humans. HEV vims-like particles (HEV VLPs) are capsid protein ieosahedral cages that can be produced by expression of the major capsid protein HEV Open Reading Frame 2 (ORF2) in a eukaryotic expression system. HEV VLPs are stable in acid and proteolytic
environments, a feature that is required for the natural transmission route of HEV. Thus, HEV VLPs represent a promising nano-carrier that can be exploited, e.g., for
chemotherapeutic delivery, vaccination, and/or imaging.
[0004] However, in order to fulfill their promise as nano-carriers, there remains a need to develop HEV VLPs that retain their stability in acid and proteolytic environments, as well as exhibit altered VLP-surface functionality. For example, there remains a need to develop HEV VLPs that can be directed to target ceils or tissue-types through surface exposed targeting moieties. The present invention fulfills this and other related needs. BRIEF SUMMARY OF THE INVENTION
[0005] The present invention provides a cysteine modified HEV V LP scaffold for altering HEV VLP surface functionality. [0006] In a first aspect, the present invention provides a modified capsid protein comprising a portion of hepatitis E virus (HEV) open Reading Frame 2 (ORF2) protein that is able to form an acid and proteolyticaliy stable HEV virus like particle (VLP), wherein: the portion of HEV ORF2 comprises a P-domain of the HEV ORF2 protein; the P-domain comprises at least one surface variable loop and a C -terminus; the P-domain comprises a cysteine in the at least one surface variable loop or at the C-termmus; and the HEV ORF2 portion retains its ability to form an acid and proteolyticaliy stable HEV VLP when the surface variable loop or C-terminal cysteine is chemically derivatized.
[0007] In some embodiments, the HEV ORF2 portion retains its ability to form an acid and proteolyticaliy stable HEV VLP when the surface variable loop or C-terminal cysteine is alkylated, acylated, arylated, succinylated, oxidized, or conjugated to a detectable label or bioactive agent. In some embodiments, the modified capsid protein comprises an amino acid sequence at least 90%, 95%, or 99% identical, or identical, to residues 112-608 of the HEV ORE 2 protein of SEQ ID NO: 1, 2, 3, 4, 5, or 6. In some embodiments, the at least one P- domain surface variable loop or C-terminal cysteine is conjugated to a detectable label. In some embodiments, the detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20. In some embodiments, the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanocluster. [0Θ08| In some embodiments, the at least one P-domain surface variable loop or C-terminai cysteine is conjugated to a bioactive agent. In some embodiments, the bioactive agent is a heterologous peptide. In some embodiments, the heterologous peptide is a ceil targeting ligand. In some embodiments, the cell targeting ligand is a cancer cell targeting liganc!. In some embodiments, the cancer ceil targeting ligand is LXY30. In some embodiments, the cancer cell targeting ligand is an antibody that binds an antigen expressed on the surface of a cancer cell. In some embodiments, the modified capsid protein further comprises a second cysteine in a P-domain surface variable loop or at the C-termimis of the P-domam. In some embodiments, the second cysteine is conjugated to a chemotherapeutic.
[Θ009] In some embodiments, the second cysteine is conjugated to a detectable label. In some embodiments, the detectable label conjugated to the second cysteine comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomi c number greater than 20. In some embodiments, the detectable label conjugated to the second cysteine comprising the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanoc luster. [0Θ10] In some embodiments, the at least one P-domain surface variable loop cysteine is alkylated, acylated, arylated, succinylated, or oxidized. In some embodiments, the at least one P-domain surface variable loop cysteine of HEV ORF2 replaces Y 85, T489, S533, N573, or T586 of HEV ORF2 or the C-terminal cysteine replaces residue 608 of HEV O F2. In some embodiments, the modified capsid protein is a component of an acid and
proteolytically stable HEV VLP.
[Θ0111 In some embodiments, the acid and proteolytically stable HEV V LP encapsulates a bioactive agent. In some embodiments, the encapsulated bioactive agent is a heterologous nucleic acid, a heterologous peptide, a detectable label, a non-protemogenic amino acid, an oligosaccharide, a synthetic macromolecule, or a chemotherapeutic. In some embodiments, the encapsulated detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20. In some embodiments, the encapsulated detectable label comprising the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanocluster. [0012] In a second aspect, the present invention provides a composition comprising the modified capsid protein of any one of the foregoing aspects or embodiments and a pharmaceutically acceptable excipient. In some embodiments, the composition comprises an HEV VLP having at least one cysteine within an HEV ORF2 P-domain surface variable loop or C-terminus that is chemically conjugated to a cell targeting ligand, a bioactive agent, or a detectable label. The chemical conjugation can result in a heterologous arrangement between the cysteine and the cell targeting ligand, bioactive agent, or detectable label. [00131 In a third aspect, the present invention provides a nucleic acid (e.g., isolated nucleic acid) comprising a polynucleotide sequence encoding any one of the foregoing modified capsid proteins. In a fourth aspect, the present invention provides an expression cassette comprising a promoter (e.g., heterologous promoter) operably linked to a polynucleotide sequence encoding an one of the foregoing modi fied capsid proteins. In a fourth aspect, the present invention provides a cell (e.g. , isolated cell or host cell) comprising the foregoing nucleic acid or the foregoing expression cassette. In a fifth aspect, the present invention provides a cell (e.g., isolated cell or host cell) comprising any one of the foregoing modified capsid proteins. In a sixt aspect, the present invention provides an organism comprising any one of the foregoing modified capsid proteins.
[0014] In a seventh aspect, the present invention provides a method of producing a modified capsid protein comprising cultivating any one of the foregoing cells, isolated cells, or host cells under conditions suitable to permit expression of the modified capsid protein. In some embodiments, the method further comprises purifying the capsid protein. In some embodiments, the method further comprises derivatizing the at least one P-domain surface variable loop or C-terminal cysteine. In some embodiments, the derivatizing comprises acylating, alkylating, arylating, succinylating, or oxidizing the P-domain surface variable loop or C-terminal cysteine. In some embodiments, the derivatizing comprises conjugating a bioactive agent to the at least one P-domain surface variable loop or C-terminal cysteine. [0015J In an eighth aspect, the present invention provides a method of directing an HE V VLP to a target cell comprising contacting a cell with the HEV-VLP, wherein the HEV VLP comprises any one of the foregoing modified capsid proteins, wherein the HEV VLP further comprises a cell targeting moiety having affinity for the target cell conjugated to the at least one P-domain surface variable loop or C-terminal cysteine, in some embodiments, the HE V VLP further comprises a detectable label, and the method further comprises detecting the detectable label. In some embodiments, the detecting the detectable label comprises detecting a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20. In some embodiments, the detecting the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises detecting a gold nanocluster.
[001 ] In some embodiments, the cell targeting moiety having affinity for the target cell conjugated to the at least one P-domain surface variable loop or C-terminal cysteine is a heterologous peptide comprising a cancer cell targeting ligarsd, and the target cell is a cancer cell. In some embodiments, the HEV VLP or a component thereof enters the cancer cell. In some embodiments, the HEV V I J' encapsulates a hioactive agent, and the method comprises delivering the bioactive agent to the intracellular region of the cell. In some embodiments, the encapsulated bioactive agent is a heterologous nucleic acid, a heterologous peptide, a detectable label, a non-proteinogenic amino acid, an oligosaccharide, a synthetic
macromolecule, or a chemotherapeutic. In some embodiments, the encapsulated detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20. In some embodiments, the encapsulated cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises an encapsulated gold nanocluster.
BRIEF DESCRIPTION OF THE DRAWINGS
[0017] Figure 1: (A) Depicts the location of surface accessible flexible coil regions (P- domain surface variable loops) of HEV ORF 2, and residues Y485, T489, S533, N573, and T586, which lie within such flexible coil regions and were selected for cysteine mutation. (B) All the selected residues for cysteine mutation were located at the surface of the P- domain, with residue Y485, T489, T586 on the outmost surface, and residues S533 and N573 at side facing the VLP 5-fold axis.
[Θ018] Figure 2: Characterization of chimeric HEV VLP carrying a cysteine replacement mutation. (A) The cysteine mutation at 573 appeared to allow efficient biotinylation of the chimeric VLP as compared to the other mutation sites, i.e., Y485, T489, S533, and T586, although those mutations exhibited similar biotinylation after VLP disassembly, (B) Both the wild type VLP and the chimeric VLP had similar binding affinity to monoclonal antibody HEP40-4B, while chimeric VLP exhibited decreased reactivity to monoclonal antibody HEP230.
[Θ019] Figure 3: (A) The three-dimensional density map of HEV wt-VLP in complex with Fab fragments of HEP230 is displayed as isosurface along a 5-fold axis, showing extra density (arrow) in the gap of two 5-fold related protruding dimers. (B) The density agreed with the footprin t of the Fab molecule. (C) Residue N573 (sphere mode) is located next to the binding site of HEP230 that (D) allows only one Fab molecule binding per 5-fold region. [0020] Figure 4: The schematic of the two-steps chemical conjugation process including a thiol selective reaction and a Copper Catalyzed Azide-Alkyne Cycloaddition (CuAAC) or "click chemistry" reaction to form LXY30-VLPs. Two methods have been tested in this research. Method 1 : (A) The Copper Catalyzed Azide-Alkyne Cycloaddition (CuAAC) reaction between Mal-Azide and Alkyne-LXY3() to form maleimide linked LXY30 (Mat- LXY30) was done first. (B) Mal-LXY30 was then added to react with Cys of N573C-VLPs. The LXY30 and Cy5.5 decorated N573C-VLP (LXY30-VLP-Cy5.5) remained intact after all these chemical conjugation processes. Method 2: (A) The conjugation process starting by labeling maleimide linked azide (Mal-Azide) at Cys sites of N573C-VLPs through Thiol selective-reaction to build azide linked VLPs (Azide-VLPs), followed by adding LXY30- Alkyne, ascorbic acid and CuS04 to form LXY30 linked VLPs (LXY30-VLPs). (C) Damaged or dis-assembled LXY30-VLPs resulting from the conjugation process, due to the presence of reactive agents including azide, ascorbic acid and CuS04, can be separated from intact VLPs by, e.g., centrifugation, size exclusion chromatography, and the like. [ΘΘ21] Figure 5: In vitro fluorescence data of VLP-Cy5.5 and LXY30-VLP-Cy5.5 binding to MDA-MB-231 breast cancer ceils. (A) The flow cytometry data of VLP-Cy5.5 or LXY30- VLP-Cy5.5 incubated with MDA-MB-231 breast cancer cells. (B) NI fluorescence images ofLXY30-Cy5.5-VLP targeting to MDA-MB-231 cells. The signal of nuclei, which were stained by Hoechst 33342 (blue), and the signal of Cy5.5 (red) were acquired using Zeiss confocal fluorescence microscopy.
[0Θ22] Figure 6: The LXY30 chemical structure and its binding to U-87MG cells. Both scrambled LXY30 and LXY30 were directly conjugated to FITC, incubated with U-87MG cells, and detected by flow cytometry. Compared with the negative control, LXY30 showed obvious binding to U-87MG cells, which highly express alpha3-betal-integrin. The scrambled LXY30 showed minimum uptake due to non-specific staining from the fluorophore FiTC.
[0023] Figure 7: In vivo NIR fluorescence images of real-time tumor targeting
characteristics ofVLP-Cy5.5 and LXY30-VLP-Cy5.5 nanoparticles on nude mice implanted with xenografts. The MDA-MB-231 breast cancer tumor bearing mice were injected i.v. with the equivalent amount of VLP-Cy5.5 or LXY30-VLP-Cy5.5. Arrow bars denote tumor sites. Optical imaging was obtained using a Kodak multimodal imaging system IS2000MM equipped with an excitation bandpass filter at 625 nm and an emission at 700 nm. [00241 Figure 8: Depicts primers designed to introduce cysteine mutations in P-domain surface variable loops of HEV ORF 2. Parental sequences with the replaced amino acid sequence boided. Forward 5 '-3' and Reverse 3 '-5 ' primers were designed with a cysteine coding sequence to the replace parental amino acid, a-e represent Y485C, T489C, S533C, N573C, and T586C respectively.
[0025 J Figure 9: Depicts the location of the surface variable loops and C -terminal residues of the P-domain of the HEV ORF2 capsid protein. (A) Depicts a schematic diagram of P- domain surface variable loops of HEV ORF 2 capsid protein. (B) Depicts the location of the P-domain surface variable loop residues and C-terminal residue on an HEV VLP. One or more cysteines can be positioned within the indicated loop or C-terminal residues for conjugation to a molecular payload.
[0026] Figure 10: Schematic depicting the application of chimeric HEV ORF2 VLPs as nanocarriers for drug delivery. The periodicity of the capsid, the presence of surface- accessible amino acids with reactive moieties, and the tolerance of a single chain version of the capsid protein dimer to diverse peptide insertions enable dense, repetitive display of targeting iigands either by chemical conjugation or genetic insertion, and display of aptamers, glycoproteins, etc., by chemical conjugation. HEV ORF2 VLPs also possess relatively large interior volumes that can be loaded with a variety of materials, including DNA plasmids. In comparison to other virus-like particles, HEV ORF2 VLPs possess strong structural plasticity, with a stability switch sensitive to calcium concentration.
[0027] Figure 11: Cys-VLP conjugation to sulfur protected Auio2. (A) Depicts two methods for covalent gold conjugation to Cys-VLP. (B) Cys-VLP direct ligand exchange with Auio2(pMBA)44. (C) Cys-VLP maieimide conjugation with Au^-Ce-Maleimide. (D) Cryo-EM Cys-VLP with A io2(pMBA) 4 a d the processed 3-D reconstruction, [0028] Figure ί 2: Schematic depicting the process used to encapsulate quantum dots in an HEV ORF2 VLP (top). Transmission electron microscope image of ferrite packaged VLPs showing darker intensity compared to package-free VLPs. DETAILED DESCRIPTION OF THE INVENTION I. Definitions
[Θ029] As used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural reference unless the context clearly dictates otherwise. [0030] "Hepatitis E virus" or "HEV" refers to a vims, virus type, or virus class, which i) causes water-borne, infectious hepatitis; it) is distinguished from hepatitis A virus (HAV), hepatitis B virus (HBV), hepatitis C vims (HCV), or hepatitis D vims (HDV) in terms of serological characteristics: and iii) contains a genomic region that is homologous to a 1.33 kb cD'NA inserted in pTZ Fl (ET1.1), a plasmid embodied in a E. coli strain deposited in American Type Culture Collection (ATCC) with accession number 67717.
[0031] The terms "capsid protein" and "modified capsid protein," with reference to HEV, refer to a mature or modified (e.g. , truncated, recombinantly mutated, or chemically derivatized) HEV open reading from 2 (ORF2) polypeptide. As used herein, reference to such ORF2 polypeptides or proteins is meant to include the full-length polypeptide, and fragments thereof, and also include any substitutions, deletions, or insertions or other modifications made to the ORF2 proteins. The capsid proteins must he capable of forming a virus like particle (VLP). Typically the capsid protein contains at least residues 112-608 of HEV ORF2, although the capsid protein can tolerate various additional substitutions, deletions, or insertions so long as they are tolerated without abrogating VLP formation. [0032] In one embodiment, the term "modified capsid protein" refers to a capsid protein, or portion thereof, in which a surface variable loop of the P-domain of HEV ORF2 is modified to incorporate one or more cysteines that are not otherwise present in the wild-type capsid protein sequence. Alternatively, or additionally, the term "modified capsid protein" refers to a capsid protein, or portion thereof, in which the C-terminus (e.g., position 608) of HEV ORF2 is modified to incorporate one or more cysteines that are not otherwise present in the wild-type capsid protein sequence. Alternatively, or additionally, the term "modified capsid protein" refers to a capsid protein, or portion thereof, in which a cysteine (e.g. , a cysteine of a surface variable loop of the P-domain of H EV ORE 2 or a cysteine recombinantly introduced at position 608) is chemically derivatized to covalentlv conjugate to the protein at least one heterologous atom or molecule. The cysteine can be inserted such that the HEV ORF2 protein length is increased, or the cysteine can replace one or more residues of a P-domain surface variable loop and/or C-terminus. [00331 Generally, modified capsid proteins retain the ability to form HEV VLPs. In some cases, the one or more cysteines are conjugated to a bioactive agent (e.g., a cell-targeting ligand such as the peptide LXY30). P-domain surface variable loops include one or more of , e.g., residues 475-493; residues 502-535; residues 539-569; residues 572-579; and residues 581-595 of HEV ORF 2 (SEQ ID NO: l, 2, 3, 4, 5, or 6). P-domain surface variable loops further include the residues of polypeptides comprising an amino acid sequence that is at least about 80%, 85%, 90%, 95%, 99%, or more identical to one or more of SEQ ID NOS:l, 2, 3, 4, 5, or 6 and that correspond to one or more of residues 475-493; residues 502-535; residues 539-569; residues 572-579; and residues 581 -595 of SEQ ID NOS: l, 2, 3, 4, 5, or 6. [0034] As used herein, the term "virus-l ike particle" (VLP) refers to an icosahedrai shell (e.g., Tl or T 3) formed by a capsid protein. VLPs are not infectious due to the lack of a viral genome. "VLP" refers to a nonreplicating icosahedrai viral shell, derived from hepatitis E vims capsid protein HE V ORF2, a portion thereof. VLPs can form spontaneously upon recombinant expression of the protein in an appropriate expression system. In some embodiments, the VLP is formed from a modified capsid protein, e.g., a capsid protein containing one or more cysteine residues in a surface variable loop of HEV ORF2, or a portion thereof. An HEV VLP can contain a mixture of modified and/or unmodified HEV ORF2 proteins.
[0035] The term "acid and proteoiytieally stable" in the context of an HEV VLP refers to an HEV VLP that is resistant to the acid and proteolytic environments of a mammalian digestive system. Methods of assessing acid and proteolytic stability are described in Jariyapong el ah, 2013, and include, but are not limited to subjecting an HEV VLP to an acid (e.g., pH of, or of about, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, or 2) and/or proteolytic environment (e.g., trypsin and/or pepsin) and examining the contacted HEV V LP by electron microscopy, gel filtration chromatography, or other suitable method to determine whether the quaternary structure (e.g., T=l, T=3, icosahedron, dodecahedron, etc.) of the HEV VLP is retained. A population of HEV VLPs (e.g., modified or unmodified) can be incubated under acid and/or proteolytic conditions for a suitable period of time (e.g., for at least, or for at least about, 1 , 2, 3, 4, 5, 10, 15, 20, 30, 45, or 60 minutes) and then tested to determine the extent of quaternary structure retention. In this context, an acid and proteoiytieally stable modified
HEV VLP refers to a modified HEV VLP that when incubated as a population of VLPs under acid and/or proteolytic conditions and assayed by electron microscopy, at least 10%, 25%, 50%, 75%, 90%, 95%, 99%, or 100% of the VLPs of the population retain their quaternary structure.
[Θ036] Alternatively, the HEV VLP can be delivered to a subject via an oral route and the efficiency of delivery assessed by detecting and/or quantifying: (i) an immune response to an antigen within the HEV VLP; (ii) a detectable label conjugated to, recombinantly introduced into, or encapsulated by the HEV V LP; or (iii) a biological response due to delivery to a cell of a bioactive agent associated with (e.g., recombinantly introduced into, conjugated to, or encapsulated by) the HEV VLP. In this context, an acid and proteolytical!y stable modified HEV VLP refers to a modified HEV VLP that retains at least 10%, 25%, 50%, 75%, 90%, 95%, 99%, or 100%) of the oral delivery efficacy and/or cell entry activity of an unmodified HEV V LP.
[0037] The term "heterologous" as used in the context of describing the relative location of two elements, refers to the two elements such as nucleic acids (e.g. , promoter or protein encoding sequence) or proteins (e.g. , an HEV ORF2 protein, or portion thereof, or modified capsid protein and another protein) that are not naturally found in the same relative positions. Thus, a "heterologous promoter" of a gene refers to a promoter that is not naturally operably linked to that gene.
[0038] Hepatitis E virus (HEV) is known to cause severe acute liver failure. HEV belongs to the genus Hepevirus in the family Hepeviridae. HEV contains a single-stranded positive- sense R.NA molecule of approximately 7.2-kb. The RNA is 3' polyadenylated and includes three open reading frames (ORE). ORF1 encodes viral nonstructural proteins, located in the 5' half of the genome. ORF2 encodes a protein-forming viral capsid, located at the 3' terminus of the genome. ORF3 encodes a 13.5-kDa protem, overlapped with C-terminus of ORF1 and N-terminus of ORF2. ORF3 is associated with the membrane as well as with the
cytoskeleton fraction.
[Θ939] The term "encapsulation," or "encapsulated," as used herein refers to the envelopment of a heterologous substance, such as a heterologous nucleic acid or protein, a chemotherapeutic, an imaging agent, a ferrite nanoparticle etc., within the VLPs defined herein. [0040] The term "bioactive agent" refers to any agent, drug, compound, or mixture thereof that targets a specific biological location (targeting agent) and/or provides some local or systemic physiological or pharmacologic effect that can be demonstrated in vivo or in vitro. Non-limiting examples include drugs, hormones, vaccines, antibodies, antibody fragments, vitamins and co factors, polysaccharides, carbohydrates, steroids, lipids, fats, proteins, peptides, polypeptides, nucleotides, oligonucleotides, polynucleotides, and nucleic acids (e.g., mRNA, tRNA, snR A, R Ai, DNA, cDNA, antisense constructs, ribozymes, etc.). [0041] A "pharmaceutically acceptable" or "pharmacologically acceptable" material is one that is not biologicall harmful or otherwise undesirable, i.e., the material may be
administered to an individual along with the capsid protein or the S UA' VLPs or the compositions of the present invention without causing any undesirable biological effects. Neither would the material interact in a deleterious manner with any of the components of the composition in which it is contained.
[0042] The term "excipient" refers to any essentially accessory substance that may be present in the finished dosage form of the composition of this invention. For example, the tenn "excipient" includes vehicles, binders, disintegrates, fillers (diluents), lubricants, glidants (flow enhancers), compression aids, colors, sweeteners, preservatives,
suspending/dispersing agents, film formers/coatings, flavors and printing inks.
[0043 j The term "adjuvant" refers to a compound that, when administered in conjunction with an antigen, augments the immune response to the antigen, but does not generate an immune response to the antigen when administered alone. Adjuvants can augment an immune response by several mechanism including lymphocyte recruitment, stimulation of B and /or T cells, and stimulation of macrophages.
[0044] An "immunogenic response" to an antigen or composition is the development in a subject of a humoral and/or a cellular immune response to an antigen present in the composition of interest. For purposes of the present disclosure, a "humoral immune response" refers to an immune response mediated by antibody molecules, while a "cellular immune response" is one mediated by T-lymphocytes and/or other white blood ceils. One important aspect of cellular immunity involves an antigen-specific response by cytolytic T- cells ("CTL"s). CTLs have specificity for peptide antigens that are presented in association with proteins encoded by the major histocompatibility complex (MHC) and expressed on the surfaces of cells. CTLs help induce and promote the destruction of intracellular microbes, or the lysis of cells infected with such microbes. Another aspect of cellular immuni ty involves an antigen- specific response by helper T-cells. Helper T-cells act to help stimulate the function, and focus the activity of, nonspecific effector cells against cells displaying peptide antigens in association with MHC molecules on their surface. A "cellular immune response" also refers to the production of cytokines, chemokines and other such molecules produced by activated T-cells and/or other white blood cells, including those derived from CD4+ and CD8+ T-cells. Hence, an immunological response may include one or more of the following effects: the production of an tibodies by B-celis; and/or the acti vation of suppressor T-celis and/or γΔ T-cells directed specifically to an antigen or antigens present in the composition or vaccine of interest. These responses ma serve to neutralize infeetivity, and/or mediate antibody-complement, or antibody dependent cell cytotoxicity (ADCC) to provide protection to an immunized host. Such responses can be determined using standard immunoassays and neutralization assays, wrell known in the art.
[0045] As used herein, the term "host" refers to humans as well as other animals.
[0046] The term "mucosal delivery" relates to delivery of a composition to a mucous membrane, such as the mucosa of the gastrointestinal tract (e.g., the buccal or labial mucosa) or the mucosa of the respiratory tract (e.g. , the nasal mucosa). //. Production and purification of modified capsid proteins and VLP formation
[ΘΘ47] One aspect of the invention relates to methods for production and purification of capsid proteins and VLPs derived therefrom (See, Expression and self-assembly of empty virus-like particles of hepatitis E virus. Li TC, Yamakawa Y, S uzuki K, Tatsumi M, Razak MA, Uchida T, Takeda N, Miyamura T., J Virol. 1997 Oct;71(10):7207-13. Essential elements of the capsid protein for self-assembly into empty virus-like particles of hepatitis E virus. Li TC, Takeda N, Miyamura T, Matsuura Y, Wang JC, Engvail H, Hammar L, Xing L, Cheng RH. J Virol 2005 Oct;79(20): 12999-3006. Niikura M et al, Chimeric recombinant hepatitis E virus-like particles as an oral vaccine vehicle presenting foreign epitopes.
Virology 2002; 293: 273-280). In one embodiment, the capsid proteins are modified capsid proteins and the VLPs derived therefrom are cysteine modified HEV VLPs. For example, the modified capsid proteins contain one or more cysteine residues in a surface variable loop of HEV OR 1 2. or a portion thereof.
[0048] Various expression systems can be used to express the capsid proteins of the present invention. Examples of expression systems useful for the production of vims- like particles of the present invention include, but are not limited to, bacterial expression system (e.g.,
Escherichia coli), insect cells, yeast cells and mammalian cells. Preferred expression system of the present invention includes baculovirus expression systems using insect cells. General methods, for example, for handling and preparing baculovirus vectors and baculoviral DNA, as well as insect cell cul ture procedures, are outlined in A Manual of Methods for
Baculovirus Vectors and insect Cell Culture Procedures.
[0049] The capsid proteins of the present invention can be cloned into the baculovirus vector, and used to infect appropriate host cells (see, for example, O'Reilly et al,
"Baculovirus Expression Vectors: A Lab Manual," Freeman & Co. 1992.). An insect cell line {e.g., Sf9 or Tn5) can be transformed with a transfer vector containing polynucleic acids which encodes the capsid proteins of the in venti on. Th e tran sfer vector in cludes, for example, linearized baculovirus DNA and a plasmid containing the desired polynucleotides. The host cell line may be co-transfected with the linearized baculovirus DNA and a plasmid in order to make a recombinant baculovirus.
[0050] Purification of the virus-like particles of the present invention can be carried out according to the standard technique in the arc (See, Li TC, et al ., J Virol. 1997
Oct;71(10):7207-13. Li TC, et al, J Virol. 2005 Oct;79(2Q): 12999-3006. Niikura M et al, Virology 2002; 293: 273-280). The purified VLPs are then resuspended in a suitable buffer.
[00511 In some embodiments, the modified capsid proteins or VLPs derived therefrom can be chemically conjugated to one or more bioactive agents. For example, one or more cysteine residues of the capsid proteins can be acyiated, alkylated, aryiated, succinyiated, or oxidized using methods known in the art. In some cases, the one or more cysteine residues can be conjugated using a maleimide functional group to covalentiy conjugate a bioactive agent to the thiol moiety of the cysteine, in some cases, the bioactive agent can be modified to introduce a maleimide functional group using CLICK chemistry. For example, an alkyne deri vative of the bioactive agent can be contacted with a maieimide-azide in the presence of C11SO4 and ascorbic acid to produce a maleimide bioactive agent. The maleimide can then be contacted with the one or more cysteines of the modified capsid protein to covalentiy link the two molecules. In some cases, the conjugating is performed on capsid protein that is not assembled into a VLP (e.g. , in the presence of EDTA, EGTA, and/or a reducing agent such as DTT or betamercaptoetha.no i). In some cases, the conjugating is performed on capsid protein that is assembled into a VLP. IIL Encapsulation of Bioactive Agents
[0052] Another aspect of the invention relates to the encapsulation of one or more bioactive agents in HEV virus-like particles (e.g., cysteine modified HEV VLPs) (See, DNA vaccine- encapsulated vims-like particles derived from an orally transmissible virus stimulate mucosa! and systemic immune responses by oral administration, Gene Therapy 2004. 11, 628-635. S Takamura, M Niikura, T-C Li, N Takeda, S usagawa, Y Takebe, T Miyamura and Y Yasutomi). Any standard technique in the art can be used to encapsulate a heterologous nucleic acid, protein, polypeptide, chemotherapeutic, imaging agent, nanoparticle, etc. into the VLPs of the present invention. The general proced ure involves (1) disassembling the VLPs formed by the capsid protein according to the present invention; and (2) reconstructing the VLPs in the presence of the bioactive agent. A skilled artisan would recognize that it is preferred to have purified VLPs before the encapsulation procedure. It is particularly preferred to have the VLPs depleted of, or substantially depleted of, any undesired materials (e.g., nucleic acids) before the encapsulation procedure.
[0053] Disassembly of VLPs can be carried out using any standard technique in the art. Reconstituted virus-like particle can be produced under physiological conditions (See, US Patent Publication No.: 20080131928). Often, disassembly of virus-like particles requires an agent to disrupt the assembly of VLPs, such as a reducing agent or a chelating agent (See, US Patent Publication No.: 20040152181). A skilled artisan would recognize that factors and conditions that affect assembly and disassembiy include: pH, ionic strength, posttranslational modifications of viral capsid proteins, disulfide bonds, and divalent cation bonding, among others. For example, the importance of cation bonding, specifically calcium, in maintaining virion integrity has been shown for polyomavirus (Brady et al., J. Virol, 23:717-724, 1977), and rotavirus (Gajardo et a!., J. Virol, 71 :2211-2216, 1997). Also, disulfide bonds appear to be significant for stabilizing polyomavirus (Walter et a!., Cold Spring Har Symp. Quant. Biol, 39:255-257, 1975; Brady et al., J. Virol, 23:717-724, 1977); and SV40 viruses (Christansen et al ., J. Virol, 21 : 1079-1084, 1977). Also, it is known that factors such as pH and ionic strength influence polyomavirus capsid stability, presumably by affecting electrostatic interactions (Brady et al., J. Virol, 23:717-724, 1977; Salunke et al., Cell, 46:895-904, 1986; Salunke et al., Biophys. J, 56:887-900, 1980). Also, it is known that post-translational modifications of some viral capsid proteins may affect capsid stability and assembly, e.g., glycosylation, phosphorylation, and acetyiation (Garcea et al, Proc. Natl, Acad. Sci. LISA, 80:3613-3617, 1983; Xi et al, J. Gen. Virol, 72:2981-2988, 1991). Thus, there are numerous interrelated factors which may affect capsid stability, assembly and disassembly.
[ΘΘ54] Preferably, the VLPs of the present invention is disassembled by the removal of calcium ions (See, Touze A, Coursaget P. in vitro gene transfer using human papillomavirus- like particles. Nucleic Acids Res 1998; 26: 1317-1323; Takamura et a!., DNA vaccine- encapsulated virus-like particles derived from an orally transmissible virus stimulate mucosal and systemic immune responses by oral administration. Gene Therapy 2004; 1 1 :628-635). According to the present invention, a reducing agent or a chelating agent or both are used to disassemble the VLPs. Various reducing agents can be used. Preferred embodiments of the reducing agents include, but are not limited to, dithiothreitol (DTT). Various chelating agents can be used, e.g., ethylene glycol tetraacetic acid (EGTA) or ethyl enediarninetetraacetic acid (EDTA). Examples of VLP disassembly conditions include, but are not limited to, the following: purified VLPs were disrupted by incubation of a buffer containing 50 mM Tris- HC1 (pH 7.5), 150 mM NaCf, 1 mM EGTA and 20 mM dithiothreitol for 30 minutes.
[0Θ55] A skilled artisan would also recognize that complete disassembly of the VLPs is not required, although preferred, to encapsulate a bioaciive agent. An artisan would also recognize that, on other occasions, it is preferred to have partial disassembly of the VLPs. A ccording to the present invention, the conditions for the partial disassembly of the VLPs can be controlled to still allow efficient encapsulation of a bioaciive agent. Partial disassembly of the VLPs can be achieved by treatment of VLPs with reducing agents alone (e.g., 20 mM DTT) (Sapp et al, j . Gen. Virol., 76:2407-2412, 1995.). According to the present invention, once the VLPs are disassembled completely or partially, encapsulation of a bioactive agent can be carried out by reassembling the VLPs in the presence of the bioactive agent. In some cases, it can be advantageous to utilize a bioactive agent having a net negative charge to enhance encapsulation. For example, nucleic acids have a net negative charge and can be preferentially encapsulated as compared to compounds that have a positive or neutral charge.
[Θ056] In some embodiments of the present invention, reassembly of the VLPs is achieved by re-supplementation of calcium ions to the disrupted V LPs. Alternatively, reassembly of the V LPs is achieved by removal of the reducing agents or the chelating agents. Optionally, factors such as pH and ionic strength, other factors described in the present inven tion, can be adjusted to achieve effi cient reassembly of the VLPs and efficien t encapsul ation of the bioactive agent.
[Θ057] In some embodiments, encapsulation is performed as follows: Following 30 mm of incubation at room temperature, a bioactive agent in 50 mM Tris-HCl buffer (pH 7.5) and 150 mM aCl is added to the disrupted VLP preparation . The disrupted VLP preparation is then refolded by incubation for 1 h with increasing concentrations of Ca(¾ up to a final concentration of 5 mM. VLPs are pel leted by ultracentrifugation and resuspended in 10 mM potassium-MES buffer (pH 6.2). To estimate the amounts of encapsulated agent, refolded and purified VLPs are purified from any unencapsulated bioactive agent and disrupted with EGTA (1 mM). Absorbance of the supernatant, or other suitable methods can be used for detection of the bioactive agent,
[0058 J In some embodiments, the bioactive agent or imaging agent to be encapsulated is conjugated to an encapsidation signal. For example, an RNA element corresponding to codons 35-59 of HEV open reading frame 1 is a powerful encapsidation signal, allowing specific interaction in vitro with HEV capsid protein, including truncated and/or cysteine modified versions of HEV ORF2 VLP as described herein. To use VLP as a carri er for therapeutic or imaging agents, chemical linkers (e.g., LC-SPDP or aptamer, teiodendrimers) that tag the agent {e.g., chemotherapeutic) with an HEV encapsidation signal like the foregoing RNA element can be used prior to the capsid self-assembly.
[0059] In some embodiments, a detectable label (imaging agent) is encapsulated. The detectable label can be a moiety renders a molecule to which it is attached to detectable by a variety of mechanisms including chemical, enzymatic, immunological, or radiological means. Some examples of detectable labels include fluorescent molecules (such as fluorescein, rhodamine, Texas Red, and phycoerythrin) and enzyme molecules (such as horseradish peroxidase, alkaline phosphatase, and β galactosidase) that allow detection based on fluorescence emission or a product of a chemical reaction catalyzed by the enzyme.
Radioactive labels involving various isotopes, such as 3H, 1251, 35S, 14C, or 32P, can also be attached to appropriate molecules to enable detection by any suitable methods that registers radioactivity, such as autoradiography. See, e.g., Tijssen, "Practice and Theory of Enzyme immunoassays," Laboratory Techniques in Biochemistry and Molecular Biology, Burdon and van Knippenberg Eds., Elsevier (1985), pp. 9 20. An introduction to labels, labeling procedures, and detection of labels can also be found in Polak and Van Noorden, Introduction to Immunocytochemistry, 2d Ed., Springer Verlag, NY (1997); and in Haugland, Handbook of Fluorescent Probes and Research Chemical s, a combined handbook and catalogue published by Molecular Probes, Inc. (1996). Further detectable labels include, but are not limited to, superparamagnetic labels {e.g., ferrite), contrast enhancing reagents (e.g., MR] contrast agents), atom-clusters (e.g., gold clusters), and the like. [00601 In some embodiments, a bioactive agent is encapsulated. In some cases, the bioactive agent is a chemotherapeutic. Suitable chemotherapeutics include, but are not limi ted to, cytotoxic drugs. Examples of cytotoxic drugs whi ch may be used in the present invention include: alkylating drugs, such as cyclophosphamide, ifospfamide, chlorambucil, meiphalan, busulfan, lomustine, carmustine, chlormethhine (mustine), estramustine, treosulfan, thiotepa, mitobronitol; cytotoxic antibiotics, such as doxorubicin, epirabicin, aciarubicin, idarubicin, daunorubicin, mitoxantrone (mitozantrone), bleomycin, dactinomycin and mitomycin; antimetabolites, such as methotrexate, capecitabine; cytarabine, fludarabine, cladribine, gemcitabine, fluorouracil, raltitrexed (tomudex), raercaptopurine, tegafur and tioguaninc; vinca alkaloids, such as vinblastine, vincristine, vindesine, vinoreibine and etoposide; other neoplastic drugs, such as amsacrine, altetarmine, crisantaspase, dacarbazine and temozolomide, hydroxycarbamide (hydroxyurea), pentostatin, platinum compounds including: carbopiatin, cisplatin and oxaliplatin, porfimer sodium, procarbazine, razoxane; taxanes including: docetaxel and paclitaxel ; topoisomerase I mhibitors including inotecan and topotecan, trastuzumab, and tretinoin. In some cases, one or more of the foregoing imaging agents and/or bioactive agents, or a combination thereof, can additionally or alternatively be conjugated to a cysteine (e.g., recombinantly introduced cysteine) in a P-domain surface variable loop or C-terminus vi a a thiol, linkage. In som e cases, one or more of the foregoing imaging agents and/or bioactive agents, or a combination thereof, can additionally or alternatively be conjugated to a second cysteine {e.g. , recombinantly introduced cysteine) in a P-domain surface variable loop or C-terminus via a thiol linkage.
[ΘΘ61] The size of the VLPs can vary when different constructs of the capsid protein are used. For example, the N-tenninal portion of the capsid protein can be adjusted to increase or decrease the size and encapsulation capacity of the VLPs. In some embodiments of the invention, in constructing the HEV VLP, a portion of HEV ORF 3 protein fused to the N- terminal of a portion of HEV ORF 2 proteins is utilized to adjust the size of the V LPs.
Typically, the HEV VLP is formed from a portion of HEV ORF2 having at least residues 1 12-608 of HEV ORF 2.
IV. Pharmaceutical compositions, formulations' and administration
[0062] The present invention also provides pharmaceutical compositions or physiological compositions comprising a cysteine modified HEV VLP formed by the capsid protein of the present invention. Such pharmaceutical or physiological compositions also include one or more pharmaceutically or physiologically acceptable excipients or carriers. Pharmaceutical compositions of the invention are suitable for use in a variety of drug delivery systems.
Suitable formulations for use in the present invention are found in Remington 's
Pharmaceutical Sciences, Mack Publishing Company, Philadelphia, PA, 17th ed. (1985). For a brief review of methods for drag delivery. See Langer, Science 249: 1527-1533 (1990), [0063] The compositions of the present invention can be administered to a host with an excipient. Excipients useful for the present invention include, but are not limited to, vehicles, binders, disintegrants, fillers (diluents), lubricants, giidants (flow enhancers), compression aids, colors, sweeteners, preservatives, suspending dispersing agents, film formers/coatings, flavors and printing inks. [0064] Various adjuvants can be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (baciile Calmette Guerin) and Corynebacterium parvum. Such adjuvants are also well known in the art. Further adjuvants that can be administered with the compositions of the invention include, but are not limited to, Monophosphoryl. lipid immunomodulator, AdjuVax 100a, QS 21 , QS 18,
CRL1005, Aluminum salts, MF 59, and Virosomai adjuvant technology. The adjuvants can also comprise a mixture of these substances. [0065] One advantage of the compositions and methods of the present invention is that the compositions of the present invention can be administered to stimulate immune response without an adjuvant. Therefore, the compositions of the present invention are in some cases administered to a host without an adjuvant.
[0066] Another advantage of the present invention is that the composi tions of the present inventio are suitable for oral delivery. Because mucosal surfaces are inter-connected, stimulation of one mucosal surface by an antigen can induce mucosal immunity not only on the directly stimulated surface, but also on the distant ones. For example, oral delivery of the compositions of the present invention can protect against respiratory and genital -urinary infections. The compositions of the present invention are also suitable for mucosal delivery, such as delivery to the buccal or labial mucosa or the respiratory tract mucosa, including the nasal mucosa. [00671 The pharmaceutical composi tions of the present invention can be administered by various routes, e.g. , oral, subcutaneous, transdermal, intradermal, intramuscular, intravenous, or intraperitoneal . The preferred routes of administering the pharmaceutical compositions are oral deliver}' at daily doses of about 0.01-5000 nig, preferably 5-500 mg. The appropriate dose may be administered in a single daily dose or as divided doses presented at appropriate intervals, for example as two, three, four, or more subdoses per day.
[0068] For preparing pharmaceutical compositions of the present invention, inert and pharmaceutical ly acceptable carriers are used. The pharmaceutical carrier can be either solid or liquid. Solid form preparations include, for example, powders, tablets, dispersible granules, capsules, cachets, and suppositories. A solid carrier can be one or more substances that can also act as diluents, flavoring agents, solubilizers, lubricants, suspending agents, binders, or tablet disintegrating agents; it can also be an encapsulating material .
[0069] In powders, the carrier is generally a finely divided solid that is in a mixture with the finely divided active component, e.g., a chimeric virus-like particles with an encapsulated nucleic acid. In tablets, the active ingredient (a chimeric virus-like particles with an encapsulated nucleic acid) is mixed with the carrier having the necessary binding properties in suitable proportions and compacted in the shape and size desired.
[0070] For preparing pharmaceutical compositions in the form of suppositories, a low- melting wax such as a mixture of fatty acid glycerides and cocoa butter is first melted and the active ingredient is dispersed therein by, for example, stirring. The molten homogeneous mixture is then poured into convenient-sized molds and allowed to cool and solidify.
[00711 Powders and tablets preferably contain between about 5% to about 70% by weight of the active ingredient. Suitable carriers include, for example, magnesium carbonate, magnesium stearate, talc, lactose, sugar, pectin, dextrin, starch, tragacanth, methyl cellulose, sodium carboxymethyl cellulose, a low-melting wax, cocoa butter, and the like.
[0O72J The pharmaceutical compositions can include the formulation of the active compound with encapsulating material as a carrier providing a capsule in which the active component (with or without other carriers) is surrounded by the carrier, such that the carrier is thus in association with the compound, in a similar manner, cachets can also be included. Tablets, powders, cachets, and capsules can be used as solid dosage forms suitable for oral administration. [0073J Liquid pharmaceutical compositions include, for example, solutions suitable for oral or parenteral administration, suspensions, and emulsions suitable for oral administration. Sterile water solutions of the active component (e.g., a chimeric virus-like particles with ars encapsulated nuc leic acid) or sterile solutions of the active component in solvents comprising water, buffered water, saline, PBS, ethanol, or propylene glycol are examples of liquid compositions suitable for parenteral administration. The compositions may contain pharmaceutically acceptable auxiliary substances as required to approximate physiological conditions, such as pH adjusting and buffering agents, tonicity adjusting agents, wetting agents, detergents, and the like. [0074] Sterile solutions can be prepared by suspending the active component (e.g., a chimeric virus-like particles with an encapsulated nucleic acid) in the desired solvent system, and then passing the resulting solution through a membrane filter to sterilize it or, alternatively, by dissolving the sterile compound in a previously sterilized solvent under sterile conditions. The resulting aqueous solutions may be packaged for use as is, or lyophilized, the lyophilized preparation being combined with a sterile aqueous carrier prior to administration. The pH of the preparations typically will be between 3 and 9, more preferably from 5 to 8, and most preferably from 6 to 7.
[0075] The pharmaceutical compositions of the present invention can be administered for prophylactic and/or therapeutic treatments. In therapeutic applications, compositions are administered to a patient already suffering from a condition in an amount sufficient to prevent, cure, reverse, or at least partially slow or arrest the symptoms of the condition and its complications. An amount adequate to accomplish this is defined as a "therapeutically effective dose." Amounts effective for this use will depend on the severity of the disease or condition and the weight and general state of the patient, but generally range from about 0.1 mg to about 2,000 mg of the composition per day for a 70 kg patient, with dosages of from about 5 mg to about 500 mg of the composition per day for a 70 kg patient being more commonly used.
[0076] In prophylactic applications, pharmaceutical compositions of the present invention are administered to a patient susceptible to or otherwise at risk of developing a disease or condition, in an amount sufficient to delay or prevent the onset of the symptoms. Such an amount is defined to be a "prophylactically effective dose." in this use, the precise amounts of the composition again depend on the patient's state of health and weight, but generally range from about 0.1 mg to about 2,000 mg of the inhibitor for a 70 kg patient per day, more commonly from about 5 mg to about 500 mg for a 70 kg patient per day,
[Θ077] Single or multiple administrations of the compositions can be carried out with dose levels and pattern being selected by the treating physician. In any event, the pharmaceutical formulations should provide a quantity of composition of the present invention sufficient to effectively stimulate immune response in the patient, either therapeutically or prophylatically.
[0078] All patents, patent applications, and other publications, including GenBank
Accession Numbers, cited in this application are incorporated by reference in the entirety for all purposes. V. Examples
[Θ079] The following examples are provided by way of illustration only and not by way of limitation. Those of skil l in the art wil l readily recognize a variety of non-critical parameters that could be changed or modified to yield essentially the same or similar results.
Example 1: Chemically Activatahle Viral Capsid Functionalized for Cancer
Targeting
Introduction
[0080] Virus-like particles (VLPs) have been proposed for use as nanocarriers to display foreign epitopes and/or deliver small molecules, a method for combating many diseases.1 This application relies mainly on the property of self-assembly as well as the ease of genetic modifications, to fulfill the designed application for the given VLP. Compared to genetic engineering, chemical conjugation of foreign peptides to VLP displays a significant advantage because it al lows a great variety of entities, such as peptide or oligosaccharide, to be conjugated to the surface of vims like particles in modulated and flexible manner without alteration of VLP assembly. [ 811 Hepatitis E virus (HEV) is composed of a non-enveloped icosahedral capsid, enclosing a single stranded RNA genome of 7.2 kilo bases. The major capsid protein is encoded by the second open reading frame (ORF2) and is essential not only for virus assembly, but also for immunogenicity and host interaction.6 The recombinant capsid protein (PORF2) is able to self-assemble into VLPs when expressed in insect cells after deletion of 111 amino acids from the N -terminal end and 52 amino acids from the C-terminal end.
PORF2 folds into three domains: S (shell; amino acids 1 18-317), M (middle; amino acids 318-451) and P (protruding; amino acids 452-606). ^11 Recombinant HEV-VLPs consists of 60 copies of PORF2, in which the S-domain possesses typical eight anti-parallel folding and stabilizes icosahedral shell and the P-domain protrudes as a surface spike that contributes greatly to HEV antigenicity. With three-domain modularity, HEV-VLP can be readily manipulated to, e.g., alter antigenicity, reduce the size from a T3 icosahedral particle to a Tl icosahedral particle, and/or modify the sequence at the P-domain without interfering with HEV-VLP assembly.
[0082] Like the native virus, HEV- VLP can be stable in acidic environment' ' and resistant to proteolytic digestion,13 thus it poses a great advantage as an oral delivery vehicle. In fact, oral administration of HEV- VLP elicits both systemic and mucosal immunity without eliciting tolerance, and thus can provide protective immunity against HEV challenge in non- human primates 15 Chimeric VLPs carrying a foreign epitope are able to elicit mucosal and systemic antibodies against both HEV and the foreign epitope after oral administration.14 Importantly, HEV-VLP can orally deliver plasmid DNA to the epithelial cells of the small intestine and induce antibody and cytotoxic T lymphocyte (CTL) responses against the plasmid encoded antigen.37 All of these studies have established the feasibility of utilizing HEV-VLP for mucosal delivery, in a route resembling virus native transmission,
[0083] Cancer theranostics requires direct contact of drag with pathological foci; therefore, a capsule carrying specific ligands is preferential for targeted delivery of anti-cancer reagents. Current delivery system based on liposome and/or organic polymers such as polyethylene glycol gives feasible yet low specificity cancer cell targeting. In this study, we created an HEV-VLP based theranostic capsule whose entry specificity can be defined by the VLP- conjugated targeting ligand. The modified HEV-VLP after conjugation with LXY30, a ligand peptide with a high affinity for human malignant breast tumor cells,58 showed specific targeting to breast tumor ceil both in vitro and in vivo, indicating that delivery route of viruslike particle can be manipulated to facilitate targeted delivery of chemotberapeutic drugs or siRNA to pathologic foci.
Results
Cystei e replacement, at. the surface loops of th e P-domain
[0Θ84] Aikyiation and acylation are the two traditional bioconjugation methods that are used to covaiently modify virus particles by attaching a ligand at amine group of lysine or thiol group on cysteine, respectively. HEV ORF2 naturally has four lysine residues that are
j 7 exposed on the surface of the P-domain. An external ligand will randomly couple with one or more of the four lysine residues, in order to avoid this random coupling, it was decided to genetically introduce a cysteine repl acement to the surface of the P-domain, which allowed acyiation to be used to site-specifically anchor the external ligand, while alkyiation was used for conjugation of a fluorescent dye to surface lysine residues.
[0085J Five sites for cysteine replacement from the surface variable loops of the P-domain were selected based on the known structure of HEV-VLP (Fig 1A),10 These sites were selected based on their location as well as the feasibility of sequence mutation to minimize any possible distortion of VLP assembly. As expected, individual cysteine replacement at Y485, T489, S533, N573, or T586 did not alter VLP assembly as the chimeric VLPs appeared as spherical projections of 27 nm in diameter under electron microscopy. To evaluate the availability for cysteine acyiation, we first examined the conjugation efficiency of HEV-VLP with maleimide-biotin which upon successful conjugation would induce VLP biotinylation through an irreversible reaction between the maieimide and thiol group of cysteine. Among ail five mutant VLPs, the chimeric VLP carrying cysteine replaced at N573 (N573C-VLP) showed significantly higher efficiency in Mal-biotinyiation than the other chimeric VLP, when assayed using labeled streptavidin. However, the dissembled N573C- VLP appeared no different in comparison to the others in the level of biotinylation (Fig 2A). These data suggested that N573C-VLP provides a suitable cysteine arrangement to accommodate biotin conjugation and/or subsequent detection.
Chimeric N573C VLP Do Not React with HEV Antibody
|0086] Because the P-domain carries the antigenic structure of HEV, sequence mutation may alter the immunoreactivity of HEV antibodies against the obtained chimeric VLPs. In order to get an insight into the antigenicity of the chimeric V LPs, the antigenicity of the chimeric N573C VLP to the wild type HEV- VLP in the presence or absence of biotin conjugation was compared. An ELISA plate was coated with N573C VLP without conjugation, maleimide-biotin conjugated N573C VLP, or the wild type HEV-VLP and then incubated with two monoclonal antibodies, HEP230 or HEP40-4. In contrast to HEP40-4, HEP230 exhibited strong binding for the wild type HEV-VLP, but failed to exhibit detectable binding to N573C VLP regardless of biotin conjugation (Fig 2B). The ELISA against the mixture of HEP230 and HEP40-4 showed similar trend (data not shown). This data suggested that residue N573 is important to HEP230 binding. Residue N573 is Involved in FA.B230 Binding
[Θ087] Proposed mutation sites lie in the surface variable loops of the P-domain. Residues Y 485, T489, and T586 are exposed on the outmost surface of the P-domain plateau while residue S533 and N573 are located on the stem region of the P-domain (Fig IB). In order to understand the role of residue N573 in HEP230 binding, we determined the structure ofHEV VLP in complex with the Fab of HEP230 by cryo-electron microscopy (cryoEM) and image processing.
[0088] Chimeric VLP in complex with Fab of HEP230 (Fab230) appeared to resemble the morphology ofHEV-VLP on cryo-electron micrographs. These complexes showed a homogenous size-distribution. The three dimensional reconstruction was calcul ted by superimposing icosahedral symmetry although the sixty antibody-binding sites on each VLP that were not fully occupied by Fab230. It revealed that HEV-VLP contains 30 protruding spikes, each of which extends outward along icosahedral two-fold axis (Fig 3A). In contrast with, the native HEV-VLP, these spikes appeared to connect with each other around the icosahedral fivefold axis. The connection density between the spikes was much weaker than the capsid and was not big enough to accommodate the Fab molecule. However, the size of the connection density appeared to be in good agreement with the contacting surface of the Fab molecule (Fig 3B), indicating that the Fab230 may contact VLP at this position.
[0089] We subsequently validated this finding by releasing the five-fold and three-fold symmetries from three-dimensional reconstruction, i.e., using 2-2-2 symmetry instead of 5-3- 2 symmetry during image processing. After five iterations of refinement, a single, slender densi ty was observed extending from the same site of the P-domain inwardly towards the 5- fold axis (data not shown). This helped to confirm that Fab230 binds to this region. Model docking with the crystal structure of an Fab fragment (PDB code 3RKD) of HE V antibody 8C 1 1 revealed that the extra density did cover the three contact loops in the Fab (Fig 3C).
Docking the crystal structure of ! i HV OR! '2 (PDB code 1ZZQ) into the density map revealed that residue N573 is in the binding footprint of Fab230 (Fig 3C, D), and could be in direct contact with the residues from Fa.b230.
LXY3Q Conjugated to N573C VLP Shows Cancer Cell Targeting [Θ090] To assess the targeting and diagnostic capability of N573-VLP, a breast cancer cell targeting ligand, L.XY30, was chemicall added to the surface N573C-VLPs through a cysteine-anchored melamine-alkyne. The LXY30-decorated N573C-VLPs (LXY30-VLPs) remained intact after the chemical conjugation (Fig. 4). Flow cytometry revealed a significantly higher attachment (>5 fold) for LXY30-VLPs to MDA-MB-231 breast cancer cells, compared to that for the wild type VLPs (Fig. 5A), indicating that LXY30-VLP specifically targets breast cancer cells. [0091] To determine if LXY30-VLPs enter the MDA-MB-231 cells, we examined the distribution of HEV ORF2 after incubation of LXY30-VLPs and MDA-M B-231 cells by the confocal microscopy. For this imaging, the LXY30-VLPs were labeled with the near infrared dye, Cy5.5, and incubated with MDA-MB-231 cells at 37 °C for 1 hour. As shown in Fig 5B, most Cy5.5-labeled VLP distributed around the perinuclear region of MDA-MB-231 cells, which meant the VLP was internalized into the cytoplasm. Wild type VLPs had minimal cellular uptake, whereas LXY30-VLPs exhibited significantly higher uptake in MDA-MB- 231 cells (Fig 5B). These results demonstrated that the chemical conjugation with target ligand is capable of eliciting the specific uptake of HEV - VLP into breast cancer cells.
LXY30 and scrambled LXY30 (S-LXY30) was also tested for uptake by the glioma cell line U-87MG, suggesting its use as a conjugate for HEV -VLP for targeting to glioma ceils (Fig. 6).
In Vivo Targeting of 1. XV 0 Conj gated to N573C VLPs to Breast. Tumor
Cells
[0092] Noninvasive in vivo imaging has been shown to be a powerful tool to visually monitor the delivery of therapeutic reagents. In vivo N1R. optical imaging was used to investigate the distribution of the VLPs in nude mice bearing MDA-MB-231 breast cancer xenografts as a criterion to evaluate the specificity of tumor targeting. Both Cy5.5 labeled LXY30-VLPs and the Cy5.5 labeled wild type VLPs were intravenously injected via tail vein in nude mice bearing DA-MB-23.1 xenografts, respectively. The mice were scanned with the fluorescence imager at various intervals for up to 72 hours. Both Cy5.5-labeled wild type VLPs and LXY30-VLPs distributed throughout the body of the mice immediately after the intravenous injection, and gradually accumulated into the MDA-MB-231 tumor via the enhanced permeability and retention (EPR) effect. However, the uptake rate of LXY30- VLPs in the tumor site was faster than that of wild type VLPs. The fluorescence intensity of Cy5.5-labeled LXY30-VLPs in MDAMB-231 tumor was also significantly higher than that of Cy5.5-labeled wild type V LPs at 1 and 6 hours post-injection (Fig. 7). [00931 HEV, as an enteric transmitted virus, preferentially enters hepatocytes in liver, it is not surprising that Cy5.5 labeled VLPs accumulated within one hour at the abdominal organs including liver. Ex vi vo imaging of excised organs were performed at 72 hours post injection. The remaining signal of Cy5.5 was found predominantly at the liver and kidney, however, the signal appeared weak in excised tumor (data not shown). The high uptake in liver and kidney could be explained in part as leaking of the breakdown VLPs from circulating vessels. Overall, these results indicate that Cy5.5/LXY30 linked HEV- VLPs is able to target MDA-MB-231 xenografts in vivo.
Discussion
[0094] There have been several attempts to use VLPs as a drug delivery system for diseases that require the delivery of cytotoxic drugs or specific genetic modifications, ceil tagging, or drugs that require encapsulation . VLPs provide an opportunity for tissue/cell specific approaches while minimizing the overall damage to healthy cells. Compared to other nano- delivery systems that have a limited half-life due to early degradation; HEV-VLP, like the Hepatitis E vims can maintain its structure through the low pH and proteolytic
mucosal/gastro environment and does not require cold-temperature for storage. This implies that HE V-VLPs could have a higher propensity for efficient delivery and reduced
accumulation due to particle degradation," both are critical factors for therapeutic nano- carriers. The baculovirus expression vector-based production of HEV ORF2 in
commercially-available insect High Five cells generates HEV-Cys-VLP capsid protein that is easily purified at high yield and low cost. ' 1 In contrast many other nano-delivery systems are compromised due to variable expression levels and inefficiency in production.
[Θ095] Cysteine replacement on the surface of the HEV -VLP P-domain was engineered so that accessibility to the ligands to the target cells was retained. Among all of the chimeric VLPs that were generated, N573C VLP construct appeared to show the strongest streptavidin binding on the basis of western blot analysis (Fig 2A ), although the cysteine thiol group is embedded within the surface depression at icosahedral fivefold axis. Structural analysis revealed that the distance between N573C and its five-fold related neighboring N573C is 45 A, a distance that is in a good agreement with the length of a streptavidin monomer. In fact, streptavidin is a homotetramer in solution. The streptavidin homotetramer can intercalate into the surface depression and form multivalent interaction with N573C anchored biotin molecules, which largely reduces the rate of dissociation. As a result, the avidity of streptavidin to N573C VLP is the highest although the affinity of streptavidin to a single biotin appeared to be the same as it showed by its binding to the disassembled VLPs. in contrast, the quaternary arrangement of cysteine residues in the other chimeric VLPs does not support multivalent binding of streptavidin.
[0096] The N573 residue is located within the binding footprint of Fab (Fig 3), so that mutation of N573 blocks the interaction of PORF2 and HEP2.30. However, the volume of a fivefold depression is not sufficient to s multaneously accommodate five Fab molecules, so that the structure of Fab-bound VLP can only capture the conta cting region of the Fab mo lecule after icosahedra!-average. The density is about 20% occupancy to that of the capsid, suggesting that only one Fab molecule resided at each five-fold axis. This is consistent with the non-icosahedral symmetry averaged density map. This binding site is different to our previously reported binding site for antibody HEP224, whose binding depends critically on residue Y485 of HEV ORF2.4 The reconstruction of HEP224 bound VLP revealed density of 60 Fabs at the shoulder of the P-domain wi th Fab extending away from VLP center.4 This suggests of two distinctive antigenic domains on the surface of HEV- VLP that are not overlapped with each other; in agreement with the ELISAl j and
mutagenesis19 results that were previously reported for HEV,
[ΘΘ97] In conclusion, the HEV-573C VLPs were chosen as a platform of modularized theranostic capsule because of its possibility of weakening the immunoresistance against the pre-existing HEV antibodies as well as the possibility of multi valent binding with the ligand, In our experiment, we utilized two different conjugation methods: thiol-selective conjugation between maleimide and cysteine, and amine-selective conjugation between amine group and NHS-ester. We chose thiol-selective conjugation for coupling a cancer cell targeting ligand to VLP, because of the spatial specificity provided by cysteine replacement. To avoid competition, the conjugation of dye was performed by amine-selective conjugation that gave rise to sufficient MR signal for us to detect VLP distribution in MDA-MB-231 breast cancer ceils in vitro and in vivo (in mice). Therefore, the HEV-Cys VLPs can be used as a platform for dual-functional, tagging with cancer adliesion-ligand and a detection marker, a robust candidate for cancer diagnosis.
[Θ098] Besides conjugating the cancer-ceil targeting ligand on the surface, the utilization of a modularized theranostic capsule can be expanded by using the interior space of HEV-Cys- VLPs as well. The HEV-Cys- VLP is capable of encapsulation of a variety of bioactive substances. For example, HEV-Cys- VLPs can encapsulate magnetic nano-particles such as ferrite, for both diagnosis under MR! and thermotherapy. in such cases, the magnetic nano- particies can be selectively stimulated using ultrasound or radio frequenc electromagnetic radiation to heat the ferrite particles encapsulated in the tumor targeted theroanostic capsule. " Negatively charged micro-RNA/siRNA could be also encapsulated into the interior of HEV-Cys-VLPs. ' By binding the cancer cell targeting ligand on its surface, HEV-Cys-VLP could have potential applications in cancer cell targeting gene therapy;
consequently, this would improve current treatments used in cancer patients.
Materials and Methods
Site-directed mutagenesis of the P domain of HEV ORF2
[0099] Five amino acid residues (Y 485, T489, S533, N573, and T586) of the P domain of HEV - ORF2 (Fig. 9) were individually mutated to a cysteine residue by site-directed mutagenesis using a Quick-Change site directed mutagenesis kit (Stratagene) following the manufacturer's protocols with the primers depicted in Fig. 8. The baculovirus transfer vector carrying the HEV ORF2 gene (pFastBacl/dORF2-HEV) was used as template for the site directed mutagenesis. These residues were chosen for mutagenesis on the basis of the dimer model and crucial binding site information of HEV.23 The mutated pFastBacl/dORF2-HEV p!asmids camming the site mutations Y485, T489, S533, N573, and T586 were named pFastBacl/dORF2-HEV-Y485, -T489, -S533, -N573, and -T586, respectively. The authentici ty of each mutation in these plasmids was confirmed by sequencing of both strands. Production and Purification of EV-Cys VLPs and In Vitro Disassembly
[0100] The mutated HEV ORF2 proteins were expressed in insect High Five ceils
(!nvitrogen, Carlsbad, CA, USA) using baculovirus-based expression vectors. The recombinant baculoviruses used to express the mutated HEV ORF2 proteins were generated using the Bac-to-Bac® Baculovirus Expression System (In vitro gen) on Sf9 cells (Invitrogen) and each of the pFastBacl/ HE V ORF2 plasmids described above according to the protocols supplied by the manufacturer. The Sf9 and High Five cells were maintained on ESF921 medium (Expression Systems, Davis, CA USA) and Exceli 420 medium (SAFC Biosciences, Lenexa, KS) supplemented with 2.5% heat-inactivated FBS, respectively, following standard protocols.24 The production and purification of HEV-Cys- VLPs from the various mutated POR2 constructs were conducted as described previously.7 in order to express the mutated POR2 proteins and generate VLPs, the High Five cells were inoculated with each baculovirus construct at a multiplicity of infection of 5 and cultured for 6-7 days. The VLPs were collected and purified through multiple steps of CsCl equilibrium densit gradient ltracentrifugation as described previously.' The purified VLPs were resuspersded in 10 mM potassium-MES buffer, pH6.2, and stored at 4 °C. A Mini dialysis device (Miliipore) was used in the disassembly experiments. The purified VLPs were disrupted by dialysis against buffer containing EDTA (lOmM) or/and DTT (20mM).
Transmission Electron Microscopy
[0101] The purified VLPs were loaded onto a glow-discharged, carbon -coated EM grid and stained with 2% uranyl acetate and examined under a JOEL. JEM-1230 transmission electron microscope at a magnification of 3Q.00QX. The images were recorded on a CCD camera (TVIPS Gauting, Germany).
Conjugation of Biotin to HEV-Cys VLPs
[0102] To chemically conjugate maleimide linked biotin to HEV-Cys VLPs, maieimide linked biotin (2ί)μΜ-200μ,Μ) was used to react with HEV-Cys V LPs (4μΜ-40μΜ) in 0.01 M PBS, pH7.2 at 4 °C overnight. The unbound maleimide-biotin was then removed using a 7,000 MWCO desalting column, (ZebaTM Spin Desalting Columns, Thermo Scientific).
Western Blotting
[0103] A total of 2 g of the biotin conjugated HEV-Cys ORF2 were loaded per lane on a 10% SDS-PAGE running under reducing condition. The proteins were then transferred to a polyvinyl! dene fluoride (PVDF) membrane (Miliipore Co.) for Western immuno'blotting. The PVDF membrane was first blocked with 5% skim milk and then incubated for 1 hour at room temperature with HRP conjugated streptavidin at a ratio of 1 : 10,000. The biotin- streptavidin reaction was then detected by an enhanced chemiluminescence method using an ECL kit (Amersham Biosciences, Piscataway, NJ).
Enzyme-Linked Immunosorbent Assay (EL!SA)
[0104] Both recombinant HEV -WT-VLP particles including HEV-WT and HEV-Cys mutants were prepared in 10 mM potassium-MES, pH 6.2 coating buffer. The proteins were diluted to the final concentrations of 1-100 ng/ml and coated overnight at 4 °C onto a clear bottom 96-well plate (Nunc, Pleasant Prairie, WI). Control wells were inclubated with 0.01 M PBS, pH 7.2. The coated VLPs were incubated with ~ 50 μΐ of the HEV antibodies for two hours at 37 °C after blocking with 0.5% Tween-20 in Tris buffer containing 20 mM Tris, pH 7.4, and 150 mM NaCl. The associated HEV antibodies were detected using alkaline phosphatase-labeled anti-mouse FAb. The enzymatic reaction was developed using pnitrophenyiphosphate (pNPP) solution (Sigma Co). The yellow color product of nitrophenyi was measured at 405 ra using a microplate reader and the average absorbance value of each VLP was calculated.
Preparation ofLXYSO- VLP-Cv5.5
[0105] Maleirnide-conjugated LXY30 was prepared by mixing 650 μΜ maleimide-azide and 650 μΜ aikyne-LXY~3Q in the presence of 200 μΜ CuS04 and 1 mM ascorbic acid at 4 °C overnight. Then maleirnide-conjugated LXY30 was added at molar ratio of 3: 1 (ligand vs. binding site) to react with the C s residues on HEV-Cys mutants though a thiol reaction at 4 °C overnight. The unconjugated molecules were removed by a 7,000 MWCO desalting columns, (ZebaTM Spin Desalting Columns, Thermo Scientific). [0106] To chemically conjugate Cy5.5 NHS ester (Limiprobe) to HEV-Cys VLPs, Cy5.5 NHS ester (200nm) was used to react with HEV-Cys mutants (20μΜ) at molar ratio of 300: 1 (dye vs VLP) in buffer containing 0.01 M PBS, pH 7.2 at room temp for 2 hrs, following by incubation at 4 °C overnight. The free Cy5.5 NHS ester was then removed by a 7,000 MWCO desalting column, (ZebaTM Spin Desalting Columns, Thermo Scientific). Flow Cytometry
[0107] MDA-MB-231 breast cancer cells were incubated with Cy5.5-labeled HEV-573C VLPs (VLP-Cy5.5) or Cy5.5/LXY30 conjugated HEV-573C VLPs (LXY3-VLP Cy5.5) for 1 hour at 37 °C, respectively. Then the cells were washed with PBS 3 times and re-suspended in PBS for the flow cytometric analysis.1 A total of 0, 000 events were collected for each sample. Unstained ceils were used as a control.
Confocal Microscopy
[0108] The sample presparation was performed according to previously established protocols. " Briefly, MDA-MB-231 breast cancer cells were seeded in the coverglass chamber slides (VWR LabShop, Batavia, IL). After reaching 80% confluence, the ceils were incubated with Cy5.5-!abeled HEV-573C VLPs (VLP-Cy5.5) and Cy5.5-labeled LXY30- c njugated HEV-573C VLPs (LXY3-VLP-Cy5.5), respectively for 1 h at 37 °C. The cell nuclei were stained with Hoechst 33342 (invitrogen) for 5 min. The ceils were washed twice with PBS and replaced with fresh medium before observation under a Zeiss confocal fluorescence microscope. In vivo Optical Imaging
[Θ109] Female SPF BALB/c mice, 10-12 weeks age, were purchased from Charles River (Davis, CA)) and kept under pathogen-free conditions according to AAALAC guidelines and were allowed to acclimatize for at least 4 days prior to any experiments. DA-MB-231 breast cancer cells were injected subcutaneously into nude mice to form subcutaneous nodules. All animal experiments were performed in compliance with institutional guidelines and according to protocol No. 06-12262 approved by the Animal Use and Care
Administrative Advisory Committee at the University of California, Davis. MDA-MB-231 tumors bearing mice were intravenously injected through the tail vein with 4 nmol/L Cy5.5 fluorescent-labeled HEV-573C VLPs (VLP-Cy5.5) and LXY30 conjugated HEV-573C VLPs (LXY30- VLP-Cy5.5) respectively.
[0110] At different time points (1, 6, 24 and 48 hours post-injection), the mice were scanned with Kodak imaging system IS20QQMM according to the procedure described previously." Images were collected with an excitation bandpass filter at 625 nm and an emission at 700 nm, exposure time was 30 s per image. After in vivo imaging, animals were euthanized by C02 overdose at 72 h after injection. Tumors, organs, and muscle tissue were excised and imaged with the Kodak imaging station. Preparation of VLP-Fab Complexes for Cryoelectron Microscopy
[0111] The VLP-Fab complexes were prepared by incubating VLPs and Fabs at a molar ratio of 1 : 180 at 4 °C overnight. To ensure optimal purity, maxima] occupancy, and ultimately reduced background noise during structural determination, pure VLP-FAB complexes were obtained by passing the sample through a short Sephacryl-300 gel -filtration column. Optical density readings at a wavelength of 280 nm were used to select the fractions that contained VLP-Fab complexes. The purified VLP fraction was then characterized by SDS-PAGE under reducing condition to validate the binding of Fab.
Cryoelectron M icroscopy
[Θ112] Procedures regarding cryo-EM and its sample preparation were essentially similar to as those previously described.2 '' Briefly, a drop of HEV-VLP aliquot of optimal concentration was applied to a glow-discharged holey carbon-coated copper grid, blotted to remove excess liquid, and immediately plunged into liquid ethane. The frozen-hydrated specimen was then transferred onto a Gatan 626DH cryo holder and examined under a transmission electron microscope (JEM-2100F, JEOL) at liquid nitrogen temperature.
Micrographs were acquired under minimal dose system on a TemCam-F415 CCD camera
(TVIPS) at a pixel size of 2 A at the specimen space. The applied defocus varied from 1 ,000 to 3,000 rati among the micrographs. Particle concentration, optimal ice thickness, and minimal specimen shift were the visual criteria for selecting micrographs.
Image Processing
[0113] Particles were then manually selected and initially centered via cross-correlating each one against the sum of the image circular average. Astigmatism and defocus values were evaluated by superimposing power spectra from all the particles within a single micrograph. The contrast transfer function was determined and phase correction was applied to the data used for structural determination. The initial model of the wild type HEV-VLP
I '""8 was obtained from previous study. " The Polar Fourier transformation (PFT) algorithm" was used to iteratively carry out origin and orientation refmementss for each part cle. Three- dimensional reconstructions were computed by combining a set of particles with orientations that spread evenl in an icosahedral asymmetric unit while superimposing the 5-3-2 icosahedral symmetry. The reliability of the reconstruction was assessed with classical Fourier Shall Correction and the final resolution was determined at 15 A using 0.5 as cut off. Fitting the Crystal Structure into Cryo-EM Density Maps
[0114] Manual fitting was carried out by transiationai and rotational movement of the HEV decanter composed often units of the HEV ORF2 protein (PDB ID 2ZZQ) into the cryo-EM density maps using program O.29 The fitting was first manually refined by minimizing the crashes between symmetry related HEV QRF2 molecules and then evaluated based on the cross correlation coefficient (CC value) between the cryo-EM density and the density computed from the fitted HEV ORF2 coordinates. Fitting was halted when the CC value reached 80%. The Fab coordinates were obtained from the crystal structure of FabSCl 1 bound HEV P-domain (PDB ID 3R D) and was docked into the cryoEM map while maintaining the binding interface as the contacting surface to the VLP. References
[0115] 1. Ludwig, C; Wagner, R., Vims-like particles-universal molecular toolboxes. Curr Opin Biotechno! 2007, 18, 537-545.
[0116] 2. Puree!!, R. H., Hepatitis E virus. In Fields Virology, 3rd Ed. ed.; Fields, B. ,; Knipe, D. M.; Howley, P. M., Eds. LIppincott - Raven Publishers: Philadelphia, 1996; pp 2831-2843. [0117J 3. Tarn, A. W.; Smith, . M.; Guerra, VI . E.; Huang, C. C; Bradley, D. W.; Fry, K. E.; Reyes, G. R., Hepatitis E virus (HE V); molecular cloning and sequencing of the full- length viral genome. Virology 1991 , 185 (1), 120-31.
[0118] 4. Xing, L.; Wang, J. C; Li, T. C; Yasutomi, Y.; Lara, L; Khudyakov, Y.;
Schofield, D.; Emerson, S. U.; Purcell, R. H.; Takeda, N.; Miyamura, T.; Cheng, R, H., Spatial configuration of hepatitis E virus antigenic domain. J Virol 2011 , 85 (2), 1 1 17-24.
[0119] 5. Schofield, D. J.; Glamann, 1; Emerson, S. U.; Purcell, R, H., Identification by phage display and characterization of two neutralizing chimpanzee monoclonal antibodies to the hepatitis E virus capsid protein. J Virol 2000, 74 (12), 5548-5555. [01201 6. Yu, H.; Li, 8.; Yang, C. Y.; Wei, ML; Song, C; Zheng, Z.; Gu, Y.; Du, H.; Zhang, J.; Xia, N. S., Homology model and potential virus-capsid binding site of a putative HEV receptor Grp78. J Moi Model 2010, Epub ahead of print.
[0121] 7. Li, T.-C; Takeda, N.; Miyamura, T.; Matsuura, Y.; Wang, J. C. Y.; Engval!, H.; Hammar, L.; Xing, L.; Cheng, R. H., Essential elements of the capsid protein for seif- assembly into empty virus-like particles of hepatitis E vims. J. Virol. 2005, 79 (20 ), 12999- 13006.
[0122] 8. Li, T. C; Yamakawa, Y.; Suzuki, K.; Tatsumi, M.; Razak, M. A.; Uchida, T.: Takeda, N.; Miyamura, T., Expression and self-assembly of empty virus-like particles of hepatitis E virus. J Virol 1997, 71 (10), 7207-7213. [0123] 9. Guu, T.; Liu, Z.; Ye, Q.; Mata, D.; Li, K.; Yin, C; Zhang, J.; Tao, Y., Structure of the hepatitis E virus-li ke particle suggests mechanisms for vims assembly and receptor binding. Proc Natl Acad Sc: U S A 2009, 106, 12992- 12997.
[0124] 10. Xing, L.; Li, T. C; Miyazaki, N.; Simon, M. N.; Wall, J. 8.; Moore, M.; Wang, C. Y.; Takeda, N.; Wakita, T.; Miyamura, T.; Cheng, R. H,, Structure of hepatitis E virion- sized particle reveals an RNA-dependent viral assembly pathway. J Biol Chem 2010, 285, 33175-33183.
[0125] 1 1 , Yamashita, T.; Mori, Y.; Miyazaki, N.; Cheng, H.; Yoshimura, M.; Unno, H.; Shima, R.; Moriisiii, K.; Tsukihara, T.; Li, T. C; Takeda, .; Miyamura, T.; Matsuura, Y., Biological and immunological charaeterics of hepatitis E virus-like particles based on the crystal structure. Proc Natl Acad Sci U S A 2009, 106, 12986-12991. [Θ126| 12. Xing, L„; Kato, K.; Li, T.; Takeda, N.; Miyamura, T.; Hammar, L.; Cheng, R. H., Recombinant hepatitis E capsid protein self-assembles into a dual-domain T = 1 particle presenting native vims epitopes. Virology 1999, 265 (1 ), 35-45.
[0127] 13. Jariyapong, P.; Xing, L.; van Houten, N. E.; Li, T. C; Weerachatyanukul, W.; Hsieh, B.; Moscoso, C. G.; Chen, C. C; iikura, M.; Cheng, R. H., Chimeric hepatitis E virus-like particle as a carrier for oral -deliver}'. Vaccine 2013, 31 (2), 417-24.
[0128] 14. Niikura, M.; Takamura, S.; Kim, G.; Kawai, S.; Saijo, M.; Morikawa, 8.;
Rurane, I.; Li, T. C: Takeda, N.; Yasutomi, Y., Chimeric recombinant hepatitis E virus-like particles as an oral vaccine vehicle presenting foreign epitopes. Virology 2002, 293 (2), 273-280.
[Θ129] 15. Zafrullah, M.; Khurslieed, Z.; Yadav, S.; Sahgal, D.; Jameel, S.; Ahmad, F., .Acidic pH enhances structure and structural stability of the capsid protein of hepatitis E virus. Biochem Biophys Res Commun 2004, 313 (1), 67-73.
[0130] 16. Li, T.-C; Suzaki, Y.; Ami, Y.; Dhole, T. N.; Miyamura, T.; Takeda, N., Protection of cynomoigus monkeys against HEV infection by oral administration of recombinant hepatitis E virus-like particles. Vaccine 2004, 22, 370-377.
[0131] 17. Takamura, 8.; Niikura, M.; Li, T. C; Takeda, N.; Kusagawa, S.; Takebe, Y.; Miyamura, T.; Yasutomi, Y ., DNA vaccine-encapsulated virus-like particles derived from an orally transmissible virus stimulate mucosal and systemic immune responses by oral administration. Gene Ther 2004, 11 (7), 628-635.
[0132] 1 8. Yao, N.; Xiao, W.; Wang, X.; Marik, J.; Park, S. H.; Takada, Y.; Lam, K. S., Discovery of targeting ligands for breast cancer cells using the one-bead one-compound combinatorial method. J Med Chem 2009, 52 (1), 126-33.
[0133J 19. Galaway, F. A,; Stockley, P. G., M52 viruslike particles: a robust, semisynthetic targeted drug delivery platform. Molecular pharmaceutics 2013, 10 (1), 59-68.
[0134] 20. Ma, Y.; Nolte, R. J.; Corneiissen, J. J., Virus-based nanocarriers for drug delivery. Advanced drug delivery reviews 2012, 64 (9), 811-25.
[0135] 21 , Kawano, M.; Xing, L„; Lam, K. S.; Handa, PL; Miyamura, T,; Barnett, S.;
Sreivastava, I. K.; Cheng, R. H., Design platforms of nanocapsules for human therapeutics or vaccine In Development of Vaccines Singh; Srivastava, L ., Eds. Wiley Pub 201 1 ; pp 125-140.
[Θ136] 22. Roemer, R. B., Engineering aspects of hyperthermia therapy. Annu Rev Biorned Eng 1999, 1, 347-76. [0 ! 7! 23. Nilsson, J.; Miyazaki, .; Xing, L.; Wu, B.; Hammar, L.; Li, T. C; Takeda, N.; Miyamura, T.; Cheng, R, H., Stmcture and Assembly of a T=l Virus-Like Particle in BK Poiyomavirus. J. Virol. 2005, 79 (9), 5337-5345.
[0138] 24. Merrington, C. L.; King, L. A.; R.D., P., Baculovirus expression systems. In Protein Expression: A practical Approach., Higgins, S. J.; Haines, B. D., Eds. Oxford University Press: 1999; pp 101 -127.
[0139] 25. Xiao, K.; Li, Y.; Lee, J. S.; Gonik, A. M; Dong, T.; Fung, G.; Sanchez, E.; Xing, L,; Cheng. H. R.; Luo, J.; Lam, K. S,, "OA02" peptide facilitates the precise targeting of paclitaxel-ioaded micellar nanoparticles to ovarian cancer in vivo. Cancer Res 2012, 72 (8), 2100-10. [0140] 26. Luo, J.; Xiao, K.; Li, Y.; Lee, J. S.; Shi, L.; Tan, Y.; Xing, L.; Cheng, H.; Liu, G.; Lam, . S., Wel idefined, size-tunable, mul ifunctional micelles for efficient paclitaxel delivery for cancer treatment. Bioconjugate Chem. 2010, 21, 1216-1224.
[0141] 27. Kawa.no, M. A.; Xing. L.; Tsukamoto, H.; Inoue, T.; Handa, H.; Cheng, R. H., Calcium bridge triggers capsid disassembly in the ceil entry process of simian viras 40. J Biol Chem 2009, 284 (50), 34703-12.
[0142] 28, Baker, T.; Cheng, H., A model based approach for determining orientations of biological macromolecules imaged by cxyo-electron microscopy. J. Struct. Biol. 1996, 116, 120-130.
[ΘΙ43] 29. Jones, T. A.; Zou, J. Y.; Cowan, S. W.; Kjeldgaard, M., Improved method for building protein model in electron density maps and the location of errors in these models. Acta crystallogr. Sect. A 1991 , 47, 110-1 1 . Example 2: Functionalized Nanocarriers
Introduction
[0144] Figure 10 depicts various routes for modifying the surface of HEV ORF2 VLP nanocarriers and encapsulation of detectable labels, drugs, or nucleic acid payloads. An HEV ORF2 VLP-based nanocarrier can self-assemble in vitro and encapsulate anticancer therapeutics or image reagents. With genetic engineering, cancer targeting iigands can be conveniently ligated to the surface of the carrier for cancer-specific targeted delivery.
Overall, the chimeric VLP exhibits excellent biocompacibility, high absorption of near- infrared light, and structure-dependent fluorescence quenching/emission. This nanomedicme platform allows integration of multiple imaging and therapeutic modalities for superior cancer detection and therapy.
[0145] Intratumoral delivery is a challenge task for modem medical research and is a process that comprises lots of negotiations betw een nanocarriers with cells, extracellular matrix, and the vasculature inside the tumor. Described herein is an HEV -VLP based nanocarrier for targeted tumor delivery guided by imaging the in situ distribution of VLP at the whole animal level, to the tumor interior, and ultimately down to subcellular organelles. Engineered HEV-VLPs for tumor targeting are provided by adding adhesion tag(s) to the surface of the P-domain. A genetical ly inserted a fluorescence marker can also be inserted into the VLP so that it can be used as endogenous reporter. Use of these nanoparticles demonstrate the power of correlative microscopic studies on the role it plays in the development of nanocarriers.
[Θ146] The use of nanoparticles in passive and targeted drug delivery has been dramatically increased over the past decades. Nanocarriers composed of synthetic polymers, liposomes, and dendrimers, have been well-investigated, however, there are only two FDA-approved antibody-conjugates and four FDA-approved nanoparticle-based drug delivery carriers. Virus like particle (VLP) is natural provided nanocarriers that possess several attractive features. They can be self-assembled into monodisperse, nansized particles, with defined periodicity that enables dense, repetitive display of tumor targeting agents. In addition, the V LP possesses multiple unique features that make it more attractive as potential nanocarrier. [01471 HEV VLP nanoparticles can be characterized with electron microscopy in the presence or absence of contrast enhancing reagents. Microscopic characterization of nanoparticles can be routinely performed to examine the physical properties of the nanocarriers using methods described herein, such as imaging frozen hydrated
multifunctional particles, e.g. , using contrast enhanced gold atom clusters (Fig. 1 1A).
Methods and compositions for conjugating gold clusters to cysteine thioiates include direct ligand exchange and/or maleimide cysteine conjugation (see, e.g., Ackerson ei al, Bioconjug. Chem., 21 , 214-218 (2010); Ackerson et al., Methods EnzymoL, 481 : 195-230 (2010); and Maijormaki et al, PNAS, 11 1(4): 1277~81 (2014)). The direct visualization by electron microscopy precisely reveals the size, shape, and domain arrangement of the nanoparticles, as well as the protein/drug absorption on nanoparticles, factors that can influence the overall Inactivity of the nanoparticles and the efficacy of targeted tumor delivery . The
nanoparticles can be imaged with either by cryo-electron microscopy on unstained frozen hydrated specimen or by traditional electron microscopy on negatively stained specimen. The cryoEM is well-suited to imagine the biological macromolecular complexes (Fig. 11 A- D).
[0148] Moreover, correlative microscopic techniques ca be based on these newl described conjugates to achieve the utilization of multiple imaging modalities in the examination of the same tumor specimen in order to deliver information above and beyond the capability of either modality alone. For instance, li ght and electron microscopy can be used to image from the organismal level, to the organ level, to cells, and even subcellular details. In combination with Near-Infrared (MR) whole animal imaging and/or micro-CT, correlative light and electron microscopy (CLEM) allows in vitro and in vivo imaging of VLP intratumorai distribution. With, e.g., high pressure freezing and freeze substitution, tumor diagnosis can be preserved to maintain the sufficient antigenicity of HEV-VLP for immunofluorescence labeling.
[01491 Region identification is one of the critical issues for tumor diagnosis, where both light and electron microscopies indeed image the same object. For instance, with HEV VLPs described herein, the coordinates of 1 ) photoconversion of DAB (3,3 '-diammobenzidine tetrahydrochrloride); 2) dual functional probe, FluoroNanogold; and 3) EM finder grids can be mformative in multiple modalities of tumor identification. The first method uses free oxygen radicals that formed upon illumination of fluorochromes to induce oxidation of DAB in situ. While visualizing specimen under fluorescence microscope in the presence of DAB solution, the oxidized DAB forms networks that will be stained with osmium tetroxide as fine granular, dense precipitates at the sites of the former fluorescent signals. FluoroNanogold conjugated antibody is a commercially available reagent that provides both light and electron microscope contrast by conjugation of 1 ,4 nm gold clusters either with green or with red fluorophore. Unlike colloidal gold of other sizes, the 1.4 nm nanogold cluster does not quench fluorescence.
[0150] HEV VLPs described herein allow detection and visualization by 3D electron tomography to monitoring drug delivery. Electron tomography provides 3D spatial information to describe the biodistribution of drug delivery vehicles inside the tumor. In a three-dimensional entity, the microenvironment of tumor can be much more complex than what found in vitro with cultured monolayer cells. Electron tomography can be used as a complement to C LEM in analyzing the intratumoral accumulation of drug- loaded V LP, so as to elucidate the mechanism for VLP extravasation and passing through the cell layers inside a tumor.
[0151] Furthermore, besides the gold clusters, HEV VLPs can be equipped and encapsulated with additional imaging agents, e.g., water-soluble CdSe/ZnS quantum dots or Ferrite, in a disassembly-and-reassembly process (Fig 12). Quantum dots can provide an dual-functional probe to track VLP tumor uptake with correlative light and electron microscopy. The efficiency of packaging can be optimized to have efficient encapsulation, e.g., by coating the quantum dots with an HEV encapsidation signal through LC-SPDP- mediated crosslinking. An RNA element occupying codons 35-59 of HEV open reading frame 1 is a powerful encapsidation signal, allowing specific interaction in vitro with HEV capsid protein, including truncated and/or cysteine modi fied versions of HEV ORF2 VLP as described herein. To use VLP as drag carrier, chemical linkers (e.g., LC-SPDP or aptamer, telodendrimers) that tag the drug (e.g., chemotherapeutic) with an HEV encapsidation signal like the foregoing RNA element can be used prior to the capsid self-assembly.
Informal Sequence Listio
SEQ ID O: l> HEV genotype 1 (NPJ556788.1)
1 rnrprpillll Imflprnlpap ppgqpsgrrr grraggsggg f gdradsqp faipyihptn
61 pfapdvtaaa gagprvrqpa rplgsa rdq aqrpaaaarr rpttagaapl tavapahdtp
121 pvpdvdsrga ilrrqynlst spltssvatg trilvlyaapl spllplqdgt nthimateas
181 nyagyrwra iryrplvpn avggyaisis fwpqt.tt.tpt svdmnsitst dvrilvqpgi
241 asehvipser Ihyrnqgw s vetsqvaeee atsgl vmlci hgslvnsytn tpytgalgll
301 dfalelefrn ltpgn ntrv srysstarhr Irrgadgtae ltttaatrfra kdlyftstng
361 vgeigrgial tlfnladtll gglpteliss aggqlf srp wsangept klytsvenaq
421 qdkgiaiphd idlgesr vi qdydnqheqd rptpspapsr pfsvlrandv l lsltaaey
481 dqstygsstg pvyvsdsvtl vnvat.gaqav arsldwtkvt Idgrplsttq qyskt.ffvlp
541 Irgkisfwea gttkagypyn yn tasdql 1 venaaghrva istyttslga qpvsisavav
601 laphsalall edtmdypara htfddfcpec rplglqgcaf qstvaelqrl kmkvgktrel SEQ ID NO:2> HEV genotype 3 (BAH 10873,1)
1 mrpravlllf fvllpmlpap pagqpsgrrr grrsggaggg fwgdrvdsqp falpyihptn
61. pfaadvvsqs jagarprqpp rplgsawrdq sqrpsaaprr rsapagaapl t.aispapdt.a
121 pvpdvdsrga ilrrqynLst spltssvasg t l iyaapi npl lplqdgt nthimateas
181 nyaqyrvvra tiryrplvpn avggyaisis fwpqttttpt svdmrisitst dvrilvqpgi
241 aselvipser lhyrnqgwrs vettgvaeee atsglvmlci hgspvnsytn tpytgalgll
301 dfalelefrn Itpgntntrv srytstarhr Irrgadgtae Itttaatrfm kdlhftgtng
361 vge grg ia1 tlfniadtll gglpteliss aggqlf srp wsangeptv klytsvenaq
421 qdkgitiphd idlqdsr vi qdydnqheqd rptpspapsr pfsvl andv Iwlsltaaey
481 dqttvgsstn pmyvsdtvtf vnvatgagav arsldwskvt Idgrplttig qysktfyvlp
541 irgklsfwea gttkagypyn ynttasdqil ienaaghrva istvttslga gptsisavgv
601 laphsalavl edttdypara htfddfcpec rtlglqgcaf qstiaelqrl krakvgktres
SEQ ID NO:3> HEV genotype 4 (ABE27148.1)
1 mrpravlllf f l Lpmlpap pagqpsgrrr grrsggtggg fwgd vdsqp a Ipyihpt.n
61 pfasdiptat gaga prqpa rplgsawrdq sqrpaaparr rsapagaspl ta apapdta
121 pvpdvdsrga ilrrqynlst spltstiatg tnlviyaapl spilplqdgt nthiiateas
181 nyaqyrvvra tiryrplvpn avggyaisis f pqttttpt svdmnsitst dvrilvqpgi
241 aselvipser lhyrnqgwrs vetsgvaeee atsglvmlci hgspvnsytn tpytgalgll
301 dfalelefrn 11 pgnt t rv srysssarhk lc jpdgtae ltttaatrfira kdlhftgtng
361 vgevgrgial l I ladtil gglpteli ss aggql fysrp wsangeptv klytsvenaq
421 qdkgiaiphd idlgesrvvi qdydnqheqd rptpspaps pf svlrandv Iwlsltaaey
481 dqttygsstn pmyvsdtvtf vnvatgtqgv srsldwskvt Idgrplttig qysktffvlp
541 Irgklsfwea gttkagypyn ynttasdqil ienapghrvc istyttnlgs gpvsisavgv
601 iaphsalaal edtvdy ara htfddfcpec ral.glqgcaf qstvaelqrl krakvgktqey
SEQ ID O:4> HEV genotype 2 (M74506.1)
i mrprplllif 11 flpmlpap ptgqpsgrrr grrsggtggg fwgdrvdsqp faipyi tn
61 pfapdvaaas gsgprirqpa rpigstwrdq aqrpsaasrr rpatagaaal tavapahdts 121 pvpdvdsrga ilrrqynlst spltssvasg tnlvlyaapl npplpiqdgt nthimateas 181 nyaqyrvara ti yrplvpn avggyaisis fwpqttttpt svdmnsitst dvrilvqpgi 241 aselvipser Ihy rnqgwrs etsgvaeee atsql mlci hgspvnsytn tpytgalgll 301 dfalelefrn ittcntntrv srysstarhs argadgtael tttaatrfn;k dlhftglngv 361 gevgrgialt ilniadtiig glptelissa ggqifysrpv vsangeptvk lytsvenaqq 421 dkgvaiphdi dlgdsr viq dydnqheqdr ptpspapsrp fsvlrandvi wlsltaaeyd 481 qstygsstgp vyisdsvtlv nvatgaqava rsldwskvtl dgrplptveq ysktffvlpl 541 rqkisfweaq ttkagypyny tasd.qil.i enaaghrva 1 styttrlgag pvaisaaavl 601 aprsalaiie dtfdypqrah tfddfcpecr ljiqgcafq st aeiqrIk vkvgktrel
SEQ ID NO:5> HEV genotype 5{BAJ771 16)
1 rnnnmflcfac gyatmrprai llllvvllpm Ipappagqss grrrgrrsgg agsgfwgdrv
61 dsqpfalpyi ptnpfasdt iaatgtgars rqsarplgsa wrdqtqrppa asrrrstptg
121 asplta apa pdt.rpvpdvd srgailrrqy nlstspltst iasgtnlvLy aaplspllpl
181 qdgtnt.hima teasnyaqyr vvratiryrp Ivpriavggya isisfwpqtt ttptsvdmns
241 itsfdvrivv qpglaselvi pserlhyrnq gwrsvetsgv aeeeatsqlv ralcihgspvn
301 sytntpytga iglldfalei efrnltpgnt ntrvsrysst arhrlhrgad gtaeltttaa
361 trfrakdlxft gsngigevgr gialtlfnla dtllgglpte lissaggqlf ysrpvvsang
421 eptvklytsv enaqqdkgia iphdidlqds rvviqdydnq heqdrptpsp apsrpfsvlr
481 vndvlwltmt. aaeyd.qt.tyg tstdpvyvsd tvtfvnvatg aqgvarsldw skvtldgrpl
541 ttiqrhskny fviplrgkls fweagttkaq ypynynttas dqilienaag hrvcistytt
601 slgsgpvsvs
SEQ ID NO:6> HEV genotype 6(BAJ61827.1) 1 mrpravlllf lmllpmlpap pagqpsgrrr grrsggsggg fwgdrvdsqp falpyihptn
61 pfasd gaqa rarqaa rplgsawrdq sqrρsasa rr rptpacjaspl ta apapdtt
121 pvpdvds ga ilr rq nlst spltstvasg tniviyaapl gpilplqdgt nthimateas
181 nyaqy v j_ra tiryr lvpn avggyaisis f pqttttpt svdmnsitst dvrilvqpgl
241 ase1iipser lhyrnqgwrs vetsgvaeee atsglvralci hgspvnsytn tpytgalgll
301 dfa elefrn 1 pgntnt rv srytstarhr Irrcjpdgtae ltttaatrfnt kdlyftjsng
361 igevgrgial tlfnladtll gglpteliss aqgqlf srp sangeptv klytsvenaq
421 qdkgiaiphe idlgdsr i qdydnqheqd rptpspapsr pf svlrvndv lwltltaaey
481 dqttygsttn pmyvsdtvtf vnvatgaqav araldwskvt fdgrplttvq qygksffvlp
541 Irgklsf a gtvkagypyn ynt asdqil venapghrvc istyttnlgs gpvsisavgv
601 laphaataal
SEQ ID NO:7> HEV genotype 1 (NP..056788,1), residues 112-608
112 - avapahdtp pvpdvdsrga ilrrqynlsi spltssvaig tniviyaapl spl!p!qdgi nthimateas -180 181 - nyaqyrwra tiryrplvpn avggyaisis Jwpqttttpt svdmnsitst dvrilvqpgi asehvipser -250
251 - lhyrnqgwrs vetsgvaeee atsglvmlci hgsivnsytn tpytgaigil dfaieiefrn Itpgntntrv -320
321 - srysstarhr Irrgadgtae Itttaatrfm kd!yftstng vgeigrgial tifniadt!! gglpteliss -390
391 - aggqlfysrp vsangeptv klytsvenaq qdkgiaiphd idlgesrvvi qdydnqheqd rptpspapsr -
460
461 - pfsvlrandv Iwlsltaaey dqstygsstg pvyvsdsvti vnvatgaqav arsldwtkvt idgrpisttq -530
531 - qysktffvlp Irgkisfwea gttkagypyn ynttasdqil venaaghrva istyitslga gpvsisavav -600 601 laphsala -608
SEQ ID O:8> HEV genotype 3 (BAH10873.1), residues 112-608
112 - aispapd a pvpdvdsrga ilrrqynlst spltssvasg tniviyaapl npilplqdgt nthimateas -180
181 - nyaqyrwra tiryrplvpn avggyaisis fwpqttttpt svdmnsitst dvrilvqpgi aseivipser -250
251 - lhyrnqgwrs vettgvaeee atsglvmlci hgspvnsytn tpytgaigil dfaieiefrn Itpgntntr -320
321 - srytstarhr irrgadgtae Itttaatrfm kdlhftgtng vgevgrgial tlfniadtll gglpteliss -390
391- aggqlfysrp vvsangeptv klytsvenaq qdkgitiphd idlgdsrvvi qdydnqheqd rptpspapsr - 460
461- pfsvlrandv iwls!taaey dqttygsstn pmyvsdtvtf vnvatgaqav arsidvvskvt Idgrpittiq -530 531- qysktfvvlp Irgkisfwea gttkagypyn ynttasdqil ienaaghrva istyitslga gptsisavgv -600 601- laphsala -608 SEQ ID NO:9> HEV genotype 4 (ABE27148.1), residues 112-608
112- avapapdta pvpdvdsrga ilrrqynlst spltstiatg tniviyaapl spllplqdgi nthiiateas -180 181 - nyaqyrvvra tiryrplvpn avggyaisis fwpqttttpt svdmnsitst dvrilvqpgi aselvipser -250 251 - ihyrnqgwrs veisgvaeee aisglvmici hgspvnsytn tpytgalgl! dfalelefrn Itpgntntrv -320 321 - srysssarhk lcrgpdgtae Itttaatrfm kdlhftgtng vgevgrgial t!iniadt!i gglpieiiss -390 391 - aggq!fysrp vvsangeptv kiytsvenaq qdkgiaiphd idlgesrvvi qdydnqheqd rptpspapsr - 460
461 - pfsvirandv iwlsitaaey dqltygsstn pmyvsdlvtf vnvaigtqgv srsidwskvt Idgrpittiq -530 541 - qysktffvlp Irgklsfwea gitkagypyn ynttasdqil ienapghrvc istyttnigs gpvsisavgv -600 801 - iaphsa!a -608 SEQ ID NO: 10> HEV genotype 2 (M74506.1), residues 112-608
1 12- avapahdts pvpdvdsrga iirrqynist spitssvasg tnivlyaapi nppiplqdgt nthimateas -180
181- nyaqyrvara tiryrplvpn avggyaisis fwpqttttpt svdmnsitst dvrilvqpgi aseivipser -250
251- Ihyrnqgwrs veisgvaeee aisglvmici hgspvnsytn tpytgaigil dfalelefrn !ttcntntrv -320
321 - srysstarhs argadgtae! tttaatrfmk dlhftglngv gevgrgialt linladtllg glptelissa -390 391- ggqlfysrpv vsangeptvk lytsvenaqq dkgvaiphdi dlgdsrvviq dydnqheqdr ptpspapsrp -460
461- fsvlrandvl wls!taaeyd qstygsstgp v isdsvtlv nvatgaqava rsidwskvtl dgrp!ptveq -530
531- ysktffvlpl rgkisf veag ttkagypyny nttasdqili enaaghrvai siyttrlgag pvaisaaavi -600
601- aprsalal
SEQ ID NO: l 1 > HEV genotype 5(BAJ77 I 16), residues 112-608
112 srrrstptg
121 aspltavapa pdtrpvpdvd srgailrrqy nistspltst iasgtnlvly aaplspllpl
181 qdgtnthima teasnyaqyr vratiryrp Ivpnavggya isisfwpqtt ttpt svdmns
241 itstdvrivv qpglaselvi pserlhyrnq g rsvetsgv aeeeatsglv mlcr qsρ VTJ
301 sytntpytga Iglldfalel efrn.ltpg t ntrvsrysst. arhrIhrgad gtaeltttaa
361 trfrokdlxft gsngigevgr qtaltlfnla dtllgql te lissaggqlf ysrpwsang
421 eptvklytsv enaqqdkgia iphdidlgds rvviqdydnq heqdrptpsp apsrpfsvlr
481 vndvlwltmt aaeydqttyg tstdpvyvsd tvtf nvatg aqgvarsldw skvtldgrpl
541. tt iqrhskny fvl.plrqk.ls fweagt kag ypy.n nttas d.qi 1ienaaq hrvcist tt
601. slgsgpvs
SEQ ID NO: 12> HEV genotype 6(BAJ61827.1), residues 1 12-608
112 avapapdtt
121 pvpdvdsrga ii rqynist spltstvasg tnivlyaapi qpilplqdqt nthimateas
181 nyaqyrvira ti yrplvpn avggyaisis fv/pqttttpt svdmnsitst dvrilvqpgi
241 asel.iipser Ihy rnqgwrs vetsgvaeee a tsglvralci hgspvnsytn tp tgalgl 1
301 dfalelefrn Itpgntntrv s r tstarh r 1 rgpdgtae Itttaatrfm. kdlyftgsng
361 Igevgrgial tlfnladtll gglpteliss aggqif srp vvsangeptv kiytsvenaq
421 qdkgiaiphe idlgdsrvti qdydnqheqd rptpspapsr pfsvlrvndv lwltltaaey 481 dqttygsttn pmyvsdtvtf vnvatgaqgv araldwskvt fdgrplttvq qygksffvlp
541 Irgkls fwea gtvkagypyn ynttasdqiL venapqhrvc istyttnlgs qpvsisavqv 601 la haata

Claims

WHAT IS CLAIMED IS: 1. A modified capsid protein comprising a portion of hepatitis E vims (HEV) open Reading Frame 2 (ORF2) protein that is able to form an acid and proteolytically stable HEV virus like particle (VLP), wherein:
the portion of HEV ORF2 comprises a P-domain of the HEV ORF2 protein; the P-domain comprises at least one surface variable loop and a C-terminus; the P-domain comprises a cysteine in the at least one surface variable loop or at the C-terminus; and
the HEV ORF2 portion retains its ability to form an acid and proteolytically stable HEV VLP when the surface variable loop or C-terminal cysteine is chemically derivatized. 2. The modified capsid protein of claim 1, wherein the HEV GRF2 portion retains its ability to form an acid and proteolytically stable HEV VLP when the surface variable loop or C-terminal cysteine is alkylated, aeylated, arylatcd, succinylated, oxidized, or conjugated to a detectable label or bioactive agent. 3. The modified capsid protein of claim 1 or 2, wherein the modified capsid protein comprises an amino acid sequence at least 90%, 95%, or 99% identical, or identical, to residues 1 12-608 of the HEV O ! 2 protein of SEQ ID NO: l,
2,
3, 4, 5, or 6.
4. The modified capsid protein of claim 1 or 2, wherein the at least one P~ domain surface variable loop or C-terminal cysteine is conjugated to a detectable label.
5. The modified capsid protein of claim 4, wherein the detectable label comprises a fluorophore, a supeiparamagnetic label, an MRi contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
6. The modified capsid protein of claim 5, wherein the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanocluster.
7. The modified capsid protein of claim 1 or 2, wherein the at least one P~ domain surface variable loop or C-terminal cysteine is conjugated to a bioactive agent,
8. The modified capsid protein of claim 7, wherein the bioactive agent is a heterologous peptide.
9. The modified capsid protein of claim 8, wherein the heterologous peptide is a ceil targeting ligand.
10. The modified capsid protein of claim 9, wherein the cell targeting ligand is a cancer cell targeting ligand.
11. The modified capsid protein of claim 10, wherein the cancer cell targeting ligand is LXY30.
12. The modified capsid protein of claim 10, wherein the cancer cell targeting ligand is an antibody that binds an antigen expressed on the surface of a cancer cell.
13. The modified capsid protein of claim 9 or 10, wherein the modified capsid protein further comprises a second cysteine in a P-domain surface variable loop or at the C -terminus of the P-domain.
14. The modified capsid protein of claim 13, wherein the second cysteine is conjugated to a chemotherapeutic.
15. The modified capsid protein of claim 13, wherein the second cysteine is conjugated to a detectable label.
16. The modified capsid protein of claim 15, wherein the detectable label conjugated to the second cysteine comprises a fluorophore, a superparamagnetic label, an Mill contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
17. The modified capsid protein of claim 16, wherein the detectable label conjugated to the second cysteine comprising the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanocluster.
18. The modified capsid protein of claim 1 or 2, wherein the at least one P- domain surface variable loop cysteine is alkylated, acylated, arylated, succinylated, or oxidized.
19. The modified capsid protein of claim 1 or 2, wherein the at least one P- domain surface variable loop cysteine of HEV ORF2 replaces Y485, T489, S533, N573, or T586 of HEV ORF2 or the C-terminal cysteine replaces residue 608 of HEV ORF2.
20. The modified capsid protein of any one of the previous claims, wherein the modified capsid protein is a component of an acid and proteoiytically stable HEV VLP.
21. The modified capsid protein of claim 20, wherein the acid and proteoiytically stable HEV VLP encapsulates a bioactive agent.
22. The modified capsid protein of claim 21 , wherein the encapsulated bioactive agent is a heterologous nucleic acid, a heterologous peptide, a detectable label, a non-proteinogenic amino acid, an oligosaccharide, a synthetic macromolecuie, or a chemotherapeutic.
23. The modified capsid protein of claim 22, wherein the encapsulated detectable label comprises a fluorophore, a superparamagnetic label, an MRJ contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
24. The modified capsid protein of claim 23, wherein the encapsulated detectable label comprising the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises a gold nanocluster.
25. A composition comprising the modified capsid protein of any one of the previous claims and a pharmaceutically acceptable excipient.
26. The composition of claim 25, wherein the composition comprises an H EV VLP having at least one cysteine within an HEV ORF2 P-domain surface variable loop or C-terminus that is chemically conjugated to a cell targeting ligand, a bioactive agent, or a detectable label.
27. A nucleic acid comprising a polynucleotide sequence encoding the modified capsid protein of any one of claims 1 - 24.
28. An expression cassette comprising a promoter operably linked to a polynucleotide sequence encoding the modified capsid protein of any one of claims 1 - 24.
29. A cell comprising the nucleic acid of claim 27 or the expression cassette of claim 28.
30. A ceil comprising the modified capsid protein of any one of claims 1 -
31. An organism comprising the modified capsid protein of any one of claims 1 - 24.
32. A method of producing a modified capsid protein comprising cultivating the cell of claim 29 under conditions suitable to permit expression of the modified capsid protein.
33. The method of claim 32 further comprising purifying the capsid protein.
34. The method of claim 33 further comprising derivatizing the at least one P-domain surface variable loop or C-terminal cysteine.
35. The method of claim 34, wherein the derivatizing comprises acyiating, alkylating, arylating, succinylating, or oxidizing the P-domain surface variable loop or C- terminal cysteine.
36. The method of claim 34, wherein the derivatizing comprises conjugating a bioactive agent to the at least one P-domain surface variable loop or C-terminal cysteine.
37. A method of directing an HEV VLP to a target cell comprising contacting a cell with th e HEV- VLP, wherein the HEV VLP comprises any one of the modified capsid proteins of claims 1 - 24, wherein the HEV VLP further comprises a cell targeting moiety having affinity for the target cell conjugated to the at least one P-domain surface variable loop or C-terminal cysteine.
38. The method of claim 37, wherein the HEV VLP further comprises a detectable label, and the method further comprises detecting the detectable label.
39. The method of claim 38, wherein the detecting the detectable label comprises detecting a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
40. The method of claim 39, wherein the detecting the cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises detecting a gold nanocluster.
41. The method of claim 37, wherein the cell targeting moiety having affinity for the target ceil conjugated to the at least one P-domain surface variable loop or C- terminal cysteine is a heterologous peptide comprising a cancer ceil targeting ligand, and the target cell is a cancer cell.
42. The method of claim 41, wherein the HEV VLP or a component thereof enters the cancer cell.
43. The method of claim 37, wherein the HEV VLP encapsulates a bioaetive agent, and the method comprises delivering the bioactive agent to the intracellular region of the cell.
44. The method of claim 43, wherein the encapsulated bioactive agent is a heterologous nucleic acid, a heterologous peptide, a detectable label, a non-protemogenic amino acid, an oligosaccharide, a synthetic macromolecule, or a chemotherapeutic.
45. The method of claim 44, wherein the encapsulated detectable label comprises a fluorophore, a superparamagnetic label, an MRI contrast agent, a positron emitting isotope, or a cluster of elements of group 3 through 18 having an atomic number greater than 20.
46. The method of claim 45, wherein the encapsulated cluster of elements of group 3 through 18 having an atomic number greater than 20 comprises an encapsulated gold nanocluster.
EP15796952.8A 2014-05-19 2015-05-18 Chemically activated nanocapsid functionalized for cancer targeting Active EP3145540B1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201462000465P 2014-05-19 2014-05-19
PCT/US2015/031439 WO2015179321A2 (en) 2014-05-19 2015-05-18 Chemically activated nanocapsid functionalized for cancer targeting

Publications (3)

Publication Number Publication Date
EP3145540A2 true EP3145540A2 (en) 2017-03-29
EP3145540A4 EP3145540A4 (en) 2017-12-13
EP3145540B1 EP3145540B1 (en) 2020-01-29

Family

ID=54554951

Family Applications (1)

Application Number Title Priority Date Filing Date
EP15796952.8A Active EP3145540B1 (en) 2014-05-19 2015-05-18 Chemically activated nanocapsid functionalized for cancer targeting

Country Status (4)

Country Link
US (2) US10053494B2 (en)
EP (1) EP3145540B1 (en)
TW (1) TWI682937B (en)
WO (1) WO2015179321A2 (en)

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3461836A1 (en) 2017-09-28 2019-04-03 Universität zu Köln Mutated adeno-associated viral capsid proteins for chemical coupling of ligands, nanoparticles or drugs via thioether binding and production method thereof
WO2019178288A2 (en) * 2018-03-13 2019-09-19 The Regents Of The University Of California Virus-like nanocapsid for oral delivery of insulin
PL3802570T3 (en) 2018-06-06 2024-03-18 The Regents Of The University Of California Improved virus-like nanoparticles for oral delivery
CN113584189B (en) * 2021-10-08 2022-02-08 上海思路迪医学检验所有限公司 Exosome and application thereof as reference substance and standard substance

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1999004029A2 (en) * 1997-07-18 1999-01-28 The Government Of The United States Of America, Represented By The Secretary, Department Of Health And Human Services A swine hepatitis e virus and uses thereof
US7351533B2 (en) 1997-09-05 2008-04-01 Medimmune, Inc. In vitro method for disassmbly/reassembly of papillomavirus virus-like particles (VLPs). Homogeneous VLP and cavsomere compositions produced by said methods: use thereof as vehicle for improved purification, and delivery of active agents
US7005130B2 (en) 2001-01-05 2006-02-28 Virginia Tech Intellectual Properties, Inc. Avian hepatitis E virus, vaccines and methods of protecting against avian hepatitis-splenomegaly syndrome and mammalian hepatitis E
WO2006004173A1 (en) 2004-07-01 2006-01-12 The Circle For The Promotion Of Science And Engineering Viral particle-like construct and method of forming the same under physiological conditions
US8906862B2 (en) * 2009-02-27 2014-12-09 The Regents Of The University Of California Multiple antigen delivery system using hepatitis E virus-like particle

Also Published As

Publication number Publication date
US10053494B2 (en) 2018-08-21
EP3145540A4 (en) 2017-12-13
TW201546087A (en) 2015-12-16
US20170107261A1 (en) 2017-04-20
WO2015179321A2 (en) 2015-11-26
WO2015179321A3 (en) 2016-01-14
EP3145540B1 (en) 2020-01-29
TWI682937B (en) 2020-01-21
US20190031720A1 (en) 2019-01-31

Similar Documents

Publication Publication Date Title
Chung et al. Viral nanoparticles for drug delivery, imaging, immunotherapy, and theranostic applications
Zangabad et al. Nanocaged platforms: modification, drug delivery and nanotoxicity. Opening synthetic cages to release the tiger
US20190031720A1 (en) Chemically activated nanocapsid functionalized for cancer targeting
Li et al. Bionanoparticles in cancer imaging, diagnosis, and treatment
Beatty et al. Cowpea mosaic virus nanoparticles for cancer imaging and therapy
Ma et al. Virus-based nanocarriers for drug delivery
Lee et al. Adaptations of nanoscale viruses and other protein cages for medical applications
JP5686468B2 (en) Hepatitis B virus (HBV) hydrophobically modified preS-derived peptides and their use as vehicles for specifically delivering compounds to the liver
JP2013209388A (en) Nanoparticle for targeted delivery of active agent
Choi et al. Protein cage nanoparticles as delivery nanoplatforms
JP2024012529A (en) Virus-like nanocapsid for oral delivery of insulin
Yuan et al. Virus-like particle-based nanocarriers as an emerging platform for drug delivery
US20230159596A1 (en) Virus-like nanoparticles for oral delivery
Gao et al. Intracellular cargo delivery by virus capsid protein-based vehicles: From nano to micro
JP6786518B2 (en) Targeted shell for drug delivery system with carbosilane dendrimer
Stark et al. Surface modulatable nanocapsids for targeting and tracking toward nanotheranostic delivery
TWI630910B (en) Peptide-conjugated nanoparticles for targeting, imaging, and treatment of prostate cancer
Shaik et al. Artificial viruses: A nanotechnology based approach
Kotha et al. Nanomaterials Mediated Diagnosis of Lung Cancer
JP7429454B2 (en) Peptides and their uses
JP7370598B2 (en) Peptides and their uses
Koudelka et al. The use of viruses in biomedical nanotechnology
Aljabali Recent Advances in Protein-Based Nano-Templates for Drug Delivery
KR20200135224A (en) Nanocarrier with micelle structure and uses thereof
WO2009081154A1 (en) Targeted delivery of macromolecules

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20161219

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

AX Request for extension of the european patent

Extension state: BA ME

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)
A4 Supplementary search report drawn up and despatched

Effective date: 20171113

RIC1 Information provided on ipc code assigned before grant

Ipc: C07K 14/08 20060101ALI20171107BHEP

Ipc: A61K 9/50 20060101ALI20171107BHEP

Ipc: A61K 39/29 20060101AFI20171107BHEP

Ipc: A61P 37/04 20060101ALI20171107BHEP

Ipc: C07H 21/00 20060101ALI20171107BHEP

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: EXAMINATION IS IN PROGRESS

17Q First examination report despatched

Effective date: 20190314

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: GRANT OF PATENT IS INTENDED

INTG Intention to grant announced

Effective date: 20190828

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE PATENT HAS BEEN GRANTED

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 1228021

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200215

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602015046118

Country of ref document: DE

REG Reference to a national code

Ref country code: FI

Ref legal event code: FGE

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: CH

Ref legal event code: NV

Representative=s name: TR-IP CONSULTING LLC, CH

REG Reference to a national code

Ref country code: NL

Ref legal event code: MP

Effective date: 20200129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200621

Ref country code: RS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: NO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200429

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200430

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200529

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200429

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SM

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: DK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602015046118

Country of ref document: DE

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 1228021

Country of ref document: AT

Kind code of ref document: T

Effective date: 20200129

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20201030

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

REG Reference to a national code

Ref country code: BE

Ref legal event code: MM

Effective date: 20200531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200518

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20200531

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

Ref country code: AL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20200129

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20230519

Year of fee payment: 9

Ref country code: FR

Payment date: 20230525

Year of fee payment: 9

Ref country code: DE

Payment date: 20230530

Year of fee payment: 9

Ref country code: CH

Payment date: 20230610

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: SE

Payment date: 20230527

Year of fee payment: 9

Ref country code: FI

Payment date: 20230525

Year of fee payment: 9

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: GB

Payment date: 20230529

Year of fee payment: 9